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:

TREML4 receptor regulates inflammation and innate immune cell death during polymicrobial sepsis

Abstract

Sepsis is a biphasic disease characterized by an acute inflammatory response, followed by a prolonged immunosuppressive phase. Therapies aimed at controlling inflammation help to reduce the time patients with sepsis spend in intensive care units, but they do not lead to a reduction in overall mortality. Recently, the focus has been on addressing the immunosuppressive phase, often caused by apoptosis of immune cells. However, molecular triggers of these events are not yet known. Using whole-genome CRISPR screening in mice, we identified a triggering receptor expressed on myeloid cells (TREM) family receptor, TREML4, as a key regulator of inflammation and immune cell death in sepsis. Genetic ablation of Treml4 in mice demonstrated that TREML4 regulates calcium homeostasis, the inflammatory cytokine response, myeloperoxidase activation, the endoplasmic reticulum stress response and apoptotic cell death in innate immune cells, leading to an overall increase in survival rate, both during the acute and chronic phases of polymicrobial sepsis.

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: Identification and genetic ablation of Treml4.
Fig. 2: Treml4 gene ablation protects thymocytes during acute polymicrobial sepsis.
Fig. 3: TREML4 is a regulator of granulocyte homeostasis in the blood and bone marrow during acute polymicrobial sepsis.
Fig. 4: TREML4 is a regulator of neutrophil homeostasis in the blood and bone marrow during acute polymicrobial sepsis.
Fig. 5: TREML4 is a regulator of neutrophil homeostasis in the blood and bone marrow during the chronic stage of polymicrobial sepsis.
Fig. 6: Treml4 deletion improves phagocytotic clearance of P. aeruginosa infection from the lungs and improves survival in a ‘two-hit’ sepsis model.
Fig. 7: TREML4 is the regulator of serum cytokine levels during polymicrobial sepsis.
Fig. 8: TREML4 regulates apoptosis and cytokine secretion by modulating calcium flux.

Similar content being viewed by others

Data availability

The authors declare that all relevant data supporting the findings of this study are available within the paper and the Extended Data. The CRISPR screening data can be accessed from BioStudies under accession number S-BSST399. The complete RNA-seq data is available from the Sequence Read Archive (SRA) under submission number SUB7079928. The mass spectrometric data is available from the PRoteomics IDEntifications (PRIDE) database under submission number PXD019055. Source data are provided with this paper.

References

  1. Dugani, S., Veillard, J. & Kissoon, N. Reducing the global burden of sepsis. CMAJ 189, E2–E3 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  2. Hotchkiss, R. S., Monneret, G. & Payen, D. Sepsis-induced immunosuppression: from cellular dysfunctions to immunotherapy. Nat. Rev. Immunol. 13, 862–874 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Davies, R., O’Dea, K. & Gordon, A. Immune therapy in sepsis: are we ready to try again? J. Intensive Care Soc. 19, 326–344 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  4. Nedeva, C., Menassa, J. & Puthalakath, H. Sepsis: inflammation is a necessary evil. Front. Cell Dev. Biol. 7, 108 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  5. 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 

  6. Ono, S., Tsujimoto, H., Hiraki, S. & Aosasa, S. Mechanisms of sepsis-induced immunosuppression and immunological modification therapies for sepsis. Ann. Gastroenterol. Surg. 2, 351–358 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  7. Nowill, A. E. et al. Immune response resetting in ongoing sepsis. J. Immunol. 203, 1298–1312 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Boomer, J. S., Green, J. M. & Hotchkiss, R. S. The changing immune system in sepsis: is individualized immunomodulatory therapy the answer? Virulence 5, 45–56 (2014).

    Article  PubMed  Google Scholar 

  9. Zhang, H. et al. Sepsis induces hematopoietic stem cell exhaustion and myelosuppression through distinct contributions of TRIF and MYD88. Stem Cell Reports 6, 940–956 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Tzelepis, K. et al. A CRISPR dropout screen identifies genetic vulnerabilities and therapeutic targets in acute myeloid leukemia. Cell Rep. 17, 1193–1205 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Doerflinger, M., Glab, J. & Puthalakath, H. Experimental in vivo sepsis models to monitor immune cell apoptosis and survival in laboratory mice. Methods Mol. Biol. 1419, 69–81 (2016).

    Article  PubMed  Google Scholar 

  12. Hemmi, H. et al. A new triggering receptor expressed on myeloid cells (Trem) family member, Trem-like 4, binds to dead cells and is a DNAX activation protein 12-linked marker for subsets of mouse macrophages and dendritic cells. J. Immunol. 182, 1278–1286 (2009).

    Article  CAS  PubMed  Google Scholar 

  13. Hemmi, H. et al. Treml4, an Ig superfamily member, mediates presentation of several antigens to T cells in vivo, including protective immunity to HER2 protein. J. Immunol. 188, 1147–1155 (2012).

    Article  CAS  PubMed  Google Scholar 

  14. Ramirez-Ortiz, Z. G. et al. The receptor TREML4 amplifies TLR7-mediated signaling during antiviral responses and autoimmunity. Nat. Immunol. 16, 495–504 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Bouillet, P. et al. Proapoptotic Bcl-2 relative Bim required for certain apoptotic responses, leukocyte homeostasis, and to preclude autoimmunity. Science 286, 1735–1738 (1999).

    Article  CAS  PubMed  Google Scholar 

  16. Cante-Barrett, K., Gallo, E. M., Winslow, M. M. & Crabtree, G. R. Thymocyte negative selection is mediated by protein kinase C- and Ca2+-dependent transcriptional induction of Bim. J. Immunol. 176, 2299–2306 (2006); erratum 176, 3843 (2006).

  17. Caissutti, C. et al. Small fetal thymus and adverse obstetrical outcome: a systematic review and a meta-analysis. Acta Obstet. Gynecol. Scand. 97, 111–121 (2018).

    Article  PubMed  Google Scholar 

  18. Basu, S., Dewangan, S., Shukla, R. C., Anupurva, S. & Kumar, A. Thymic involution as a predictor of early-onset neonatal sepsis. Paediatr. Int. Child Health 32, 147–151 (2012).

    Article  PubMed  Google Scholar 

  19. Hatano, S. et al. Recombinant Mycobacterium bovis Bacillus Calmette-Guérin expressing Ag85B-IL-7 fusion protein enhances IL-17A-producing innate γδ T cells. Vaccine 34, 2490–2495 (2016).

    Article  CAS  PubMed  Google Scholar 

  20. Markwart, R. et al. Immunosuppression after sepsis: systemic inflammation and sepsis induce a loss of naive T-cells but no enduring cell-autonomous defects in T cell function. PLoS ONE 9, e115094 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Siracusa, F. et al. CD69+ memory T lymphocytes of the bone marrow and spleen express the signature transcripts of tissue-resident memory T lymphocytes. Eur. J. Immunol. 49, 966–968 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Luan, Y. Y., Yao, Y. M., Xiao, X. Z. & Sheng, Z. Y. Insights into the apoptotic death of immune cells in sepsis. J. Interferon Cytokine Res. 35, 17–22 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Misharin, A. V., Morales-Nebreda, L., Mutlu, G. M., Budinger, G. R. & Perlman, H. Flow cytometric analysis of macrophages and dendritic cell subsets in the mouse lung. Am. J. Respir. Cell Mol. Biol. 49, 503–510 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Roger, T. & Calandra, T. Interleukin-33 safeguards neutrophils in sepsis. Nat. Med. 16, 638–639 (2010).

    Article  CAS  PubMed  Google Scholar 

  25. Hampson, P. et al. Neutrophil dysfunction, immature granulocytes and cell-free DNA are early biomarkers of sepsis in burn-injured patients: a prospective observational cohort study. Ann. Surg. 265, 1241–1249 (2017).

    Article  PubMed  Google Scholar 

  26. Matsushima, H. et al. Neutrophil differentiation into a unique hybrid population exhibiting dual phenotype and functionality of neutrophils and dendritic cells. Blood 121, 1677–1689 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Condotta, S. A., Khan, S. H., Rai, D., Griffith, T. S. & Badovinac, V. P. Polymicrobial sepsis increases susceptibility to chronic viral infection and exacerbates CD8+ T cell exhaustion. J. Immunol. 195, 116–125 (2015).

    Article  CAS  PubMed  Google Scholar 

  28. Chen, W. et al. Ethyl pyruvate reverses development of Pseudomonas aeruginosa pneumonia during sepsis-induced immunosuppression. Int. Immunopharmacol. 52, 61–69 (2017).

    Article  CAS  PubMed  Google Scholar 

  29. Muenzer, J. T. et al. Pneumonia after cecal ligation and puncture: a clinically relevant ‘two-hit’ model of sepsis. Shock 26, 565–570 (2006).

    Article  PubMed  Google Scholar 

  30. Kremserova, S. et al. Lung neutrophilia in myeloperoxidase-deficient mice during the course of acute pulmonary inflammation. Oxid. Med. Cell. Longev. 2016, 5219056 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Galani, I. E. & Andreakos, E. Neutrophils in viral infections: current concepts and caveats. J. Leukoc. Biol. 98, 557–564 (2015).

    Article  CAS  PubMed  Google Scholar 

  32. Tate, M. D., Brooks, A. G. & Reading, P. C. The role of neutrophils in the upper and lower respiratory tract during influenza virus infection of mice. Respir. Res. 9, 57 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Seeley, J. J. et al. Induction of innate immune memory via microRNA targeting of chromatin remodelling factors. Nature 559, 114–119 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Cavaillon, J. M. & Adib-Conquy, M. Bench-to-bedside review: endotoxin tolerance as a model of leukocyte reprogramming in sepsis. Crit. Care 10, 233 (2006).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Turnbull, I. R. et al. DAP12 (KARAP) amplifies inflammation and increases mortality from endotoxemia and septic peritonitis. J. Exp. Med. 202, 363–369 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Futosi, K., Fodor, S. & Mocsai, A. Neutrophil cell surface receptors and their intracellular signal transduction pathways. Int. Immunopharmacol. 17, 638–650 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Bahar, E., Kim, H. & Yoon, H. ER stress-mediated signaling: action potential and Ca2+ as key players. Int. J. Mol. Sci. 17, 1558–1580 (2016).

    Article  PubMed Central  CAS  Google Scholar 

  38. Gorudko, I. V. et al. Neutrophil activation in response to monomeric myeloperoxidase. Biochem. Cell Biol. 96, 592–601 (2018).

    Article  CAS  PubMed  Google Scholar 

  39. Hann, J., Bueb, J. L., Tolle, F. & Brechard, S. Calcium signaling and regulation of neutrophil functions: still a long way to go. J. Leukoc. Biol. 107, 285–297 (2019).

    Article  PubMed  CAS  Google Scholar 

  40. Ayala, A., Herdon, C. D., Lehman, D. L., Ayala, C. A. & Chaudry, I. H. Differential induction of apoptosis in lymphoid tissues during sepsis: variation in onset, frequency and the nature of the mediators. Blood 87, 4261–4275 (1996).

    Article  CAS  PubMed  Google Scholar 

  41. Madsen, M. B. et al. Patient’s characteristics and outcomes in necrotising soft-tissue infections: results from a Scandinavian, multicentre, prospective cohort study. Intensive Care Med. 45, 1241–1251 (2019).

    Article  CAS  PubMed  Google Scholar 

  42. Bordon, J. et al. Understanding the roles of cytokines and neutrophil activity and neutrophil apoptosis in the protective versus deleterious inflammatory response in pneumonia. Int. J. Infect. Dis. 17, e76–e83 (2013).

    Article  CAS  PubMed  Google Scholar 

  43. Yoshida, T. et al. Relationship between neutrophil influx and oxidative stress in alveolar space in lipopolysaccharide-induced lung injury. Respir. Physiol. Neurobiol. 191, 75–83 (2014).

    Article  CAS  PubMed  Google Scholar 

  44. Delgado-Rizo, V. et al. Neutrophil extracellular traps and its implications in inflammation: an overview. Front. Immunol. 8, 81 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Warren, H. S. Mouse models to study sepsis syndrome in humans. J. Leukoc. Biol. 86, 199–201 (2009).

    Article  CAS  PubMed  Google Scholar 

  46. Stortz, J. A. et al. Murine models of sepsis and trauma: can we bridge the gap? ILAR J. 58, 90–105 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Takao, K. & Miyakawa, T. Genomic responses in mouse models greatly mimic human inflammatory diseases. Proc. Natl Acad. Sci. USA 112, 1167–1172 (2015).

    Article  CAS  PubMed  Google Scholar 

  48. Osterburg, A. R. et al. Concerns over interspecies transcriptional comparisons in mice and humans after trauma. Proc. Natl Acad. Sci. USA 110, E3370 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Feterowski, C. et al. Effects of functional Toll-like receptor-4 mutations on the immune response to human and experimental sepsis. Immunology 109, 426–431 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Klesney-Tait, J., Turnbull, I. R. & Colonna, M. The TREM receptor family and signal integration. Nat. Immunol. 7, 1266–1273 (2006).

    Article  CAS  PubMed  Google Scholar 

  51. Koike-Yusa, H., Li, Y., Tan, E. P., Velasco-Herrera Mdel, C. & Yusa, K. Genome-wide recessive genetic screening in mammalian cells with a lentiviral CRISPR-guide RNA library. Nat. Biotechnol. 32, 267–273 (2014).

    Article  CAS  PubMed  Google Scholar 

  52. Aubrey, B. J. et al. An inducible lentiviral guide RNA platform enables the identification of tumor-essential genes and tumor-promoting mutations in vivo. Cell Rep. 10, 1422–1432 (2015).

    Article  CAS  PubMed  Google Scholar 

  53. Haviernik, P., Zhang, Y. & Bunting, K. D. Retroviral transduction of murine hematopoietic stem cells. Methods Mol. Biol. 430, 229–241 (2008).

    Article  CAS  PubMed  Google Scholar 

  54. Cheng, H., Liang, P. H. & Cheng, T. Mouse hematopoietic stem cell transplantation. Methods Mol. Biol. 976, 25–35 (2013).

    Article  CAS  PubMed  Google Scholar 

  55. Droop, A. P. fqtools: an efficient software suite for modern FASTQ file manipulation. Bioinformatics 32, 1883–1884 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Manzanero, S. In Leucocytes. Methods in Molecular Biology Vol. 844 (ed. Ashman, R.) 177–181 (Humana Press, 2012).

  57. Cox, J. & Mann, M. MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification. Nat. Biotechnol. 26, 1367–1372 (2008).

    CAS  PubMed  Google Scholar 

  58. Mellacheruvu, D. et al. The CRAPome: a contaminant repository for affinity purification-mass spectrometry data. Nat. Methods 10, 730–736 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Ritchie, M. E. et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43, e47 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  60. Rappsilber, J., Mann, M. & Ishihama, Y. Protocol for micro-purification, enrichment, pre-fractionation and storage of peptides for proteomics using StageTips. Nat. Protoc. 2, 1896–1906 (2007).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank R. Anders and J. Goding for critically reading the manuscript. We thank A. Baxter for reagents, La Trobe University Animal Ethics Committee and the LARTF staff for facilitating mouse experiments and the La Trobe Bioimaging Platform for the maintenance of the flow cytometry facility. We also thank S. Wilcox and P. Hickey (Walter and Eliza Hall Institute) for RNA-seq and bioinformatics. H.P. and C.N. were supported by La Trobe University funding (RFA-UD) and the Strategic Innovation Fund. J.M. is supported by La Trobe University’s postgraduate scholarship. The generation of Treml4−/− mice used in this study was supported by the Australian Phenomics Network and the Australian Government through the National Collaborative Research Infrastructure Strategy program.

Author information

Authors and Affiliations

Authors

Contributions

H.P., C.N., J.M., M. Doerflinger and P. Fao designed the experiments. C.N., J.M., M. Doerflinger, C.L., M. Duan, P. Fon and T.K.P. conducted the experiments. A.J.K. and M.J.H. generated the Treml4−/− mouse strain. H.R. performed the bioinformatics. H.P., M. Duan, M.D.H. and W.C. analyzed the data. H.P. and C.N. prepared the manuscript.

Corresponding author

Correspondence to Hamsa Puthalakath.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Peer reviewer reports are available. L. A. Dempsey 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 HSC reconstitution.

a, Reconstitution of myelo-ablated C57BL/6 mice (CD45.1) with the donor (CD45.2) hematopoietic stem cells containing the lentiviral library before sub-lethal CS injection (these mice were used in the screen as shown in Fig. 1a,b). The level of engraftment was analysed in each of the compartments as shown in the figure. CD45.1 vs CD45.2 bone marrow populations were gated to validate recipient and donor populations pre and post- implantation into mice for initial screening experiment. b, An overlay of BFP positive bone marrow cells that were used for NGS, with control cells.

Source data

Extended Data Fig. 2 T cell phenotype during acute phase sepsis.

a, b, Thymic atrophy observed by dissection 24 h post-CS injection. c, The gating strategy for phenotyping splenic T cells during acute sepsis. d, The gating strategy for measuring apoptosis in splenic CD4 T cells during acute sepsis. e, The gating strategy for measuring apoptosis in splenic CD8 T cells during acute sepsis. f, The gating strategy for phenotyping lymph node T cells during acute sepsis. g, The gating strategy for measuring apoptosis in lymph node CD4 T cells during acute sepsis. h, The gating strategy for measuring apoptosis in lymph node CD8 T cells during acute sepsis. Gating strategy in detail: The entire splenic and LN population was gated to exclude debris using FSC-A vs SSC-A. The remaining population was further gated to exclude doublets using FSC-A vs FSC-H. T-cell exclusive population was derived using CD45.2 vs CD3. To analyse CD4 and CD8 populations specifically, further gating was performed using CD4 vs CD8. Individual populations were analysed for cell death using DAPI (necrotic cell marker) vs AnnexinV (apoptosis marker).

Extended Data Fig. 3 The bone marrow T cell phenotype during the chronic phase of sepsis.

The gating strategy and cellularity of the bone marrow CD4+ T and CD8+ T cells during the ‘two-hit’ model of pneumonia. Error bars ± SD, n = 3 mice in each group, **p < 0.005, ***p < 0.001 and #p > 0.05. Statistical significance was determined by unpaired two- tailed t-test. Gating strategy in detail: The entire bone marrow population was gated to exclude debris using FSC-A vs SSC-A. The remaining population was further gated to exclude doublets using FSC-A vs FSC-H. T-cell exclusive population was derived using CD45.2 vs CD3. To analyse CD4 and CD8 populations specifically, further gating was performed using CD4 vs CD8. Individual populations were analysed for cell death using DAPI (necrotic cell marker) vs AnnexinV (apoptosis marker).

Source data

Extended Data Fig. 4 The gating strategy for granulocytes, macrophages and dendritic cells during acute phase sepsis shown in Fig. 3.

a. Granulocyte gating strategy in the bone marrow. b, Total granulocyte cellularity in the bone marrow. Error bars ± SD, *p < 0.05, #p > 0.05 and n = 3-4 mice per group. c, Granulocyte gating strategy in the blood. d, Total granulocyte cellularity in the blood. Error bars ± SD, *p < 0.05, #p > 0.05 and n = 3-4 mice per group. e, f, The gating strategy and phenotyping of macrophages in the bone marrow. g, Cellularity of bone marrow macrophages based on the profile shown in f. Error bars ± SD, *p < 0.05, #p > 0.05 and n = 3-4 mice per group. h, Apoptosis measurement in macrophages. Error bars ± SD, n = 6-7 per group, *p < 0.05, **p < 0.005, and #p > 0.05. i, The gating strategy for splenic dendritic cells that was used in measuring splenic DC count (j), apoptosis measurement (k, m) and percentage dendritic cells (l). Error bars ± SD, n = 6–8 mice per group for j, l and m, *p < 0.05 and #p > 0.05. Statistical significance was determined by unpaired two-tailed t-test (b, d, g, h, j, l and m). Gating strategy in detail: The entire cell population within each of the tissues was gated to remove doublets using FSC-A vs FSC-H. For visualisation of the granulocyte populations in the BM and blood; CD45.2 vs FSC-A was used to isolate all leukocytes, proceeded to use CD11b vs Ly6C for phenotype analysis. For macrophage phenotype analysis in the BM; CD45.2 vs CD115 positive populations were further gated for Ly6C vs CD11b (maturation a status). For splenic dendritic cell death analysis; CD45.2 positive population was gated for MHC II (high) vs CD11c (Pan DC population), this population was further analysed for cell death using DAPI vs AnnexinV.

Source data

Extended Data Fig. 5 The gating strategy for neutrophils cells during acute phase sepsis.

a, b, The gating strategy for bone marrow neutrophils apoptosis. c, d, The gating strategy for blood neutrophils apoptosis. This gating was used for the data shown in Fig. 4. Gating strategy in detail: The entire cell population within each of the tissues was gated to remove doublets using FSC-A vs FSC-H. Neutrophils were isolated further using CD45.2 vs Ly6G. This population was further analysed for cell death using DAPI vs AnnexinV. Additional CD11b and CXCR2 markers were used to gate neutrophils for their presence and their maturation status.

Extended Data Fig. 6 Neutrophils are the regulators of cell survival during polymicrobial sepsis.

a, Treml4 deletion leads to an improved body weight upon sub-lethal injection of CS (n = 4 mice in each group). b, Treml4 deletion leads to an improved survival during lethal CS-induced polymicrobial sepsis (n = 4 mice in each group). Error bars ± SD, **p < 0.005 and ***p < 0.001. Statistical significance was determined by unpaired two-tailed t-test.

Source data

Extended Data Fig. 7 The gating strategy for analyzing the BAL neutrophils shown in Figs. 5 and 6.

Gating strategy in detail: The entire cell population within the BALwas gated to remove doublets using FSC-A vs FSC-H. Neutrophils were isolated further using CD45.2 vs Ly6G. Gating strategy in detail: The entire cell population within each of the tissues was gated to remove doublets using FSC-A vs FSC-H. Neutrophils were isolated further using CD45.2 vs Ly6G. This population was further analysed for cell death using DAPI vs AnnexinV. Additionally, CXCR2 marker was used to gate neutrophils for their presence and their maturation status.

Extended Data Fig. 8 Treml4–/– neutrophils regulate cell survival and bacterial load during polymicrobial sepsis but do not protect against viral infection.

a, Typical neutrophil depletion profile in Treml4–/– mice up on anti-Ly6G antibody injection supporting the data in Fig. 6h. b, Recovery of P. aeruginosa in the lungs of mice 24 h post-administration in a two-hit model (n = 4 in each group). c, The bacterial load in the kidney and spleen of mice 6 h post CS injection (n = 4 mice in each group). d, Survival analysis of mice challenged with influenza virus (PR8) after sublethal CS injection (n = 4 mice in each group). Error bars ± SD, **p < 0.005 and ***p < 0.001. Statistical significance was determined by unpaired two-tailed t-test (b, c) and by log rank test (d).

Source data

Extended Data Fig. 9 TREML4 receptor regulates the inflammatory pathway during polymicrobial sepsis and is independent of TLR4.

a, Time series analysis of the gene expression of wild type and Treml4–/– BMDMs, projected onto their principal components. Colours on the dots indicates time. The PCA was drawn using normalized counts from data shown in Fig. 7a. b, Protein abundance between wild type and Treml4–/– BAL neutrophils, before and after treating with CS, projected to their principal components. Colors on the dots indicate the treatment condition. The PCA was drawn using the maxLFQ values from the data presented in Fig. 8g. c, d, Raw RNASeq data for Ifn-γ and Ifn-β respectively, curated from the data presented in Fig. 7a. Statistical significance was determined by two-tailed, moderated t-test with adjustments made for multiple comparisons using Benjamini-Hochberg method.

Extended Data Fig. 10 Treml4 gene ablation does not impact TLR4 signaling.

a, b, Cytokine measurements in BMDMs and bone marrow neutrophils respectively, after challenging them with various concentrations of LPS in vitro. Error bars ± SD, n = 3 independent biological replicates and # indicates lack of any statistical significance. Statistical significance was determined by unpaired two-tailed t-test. c, Densitometric quantitation of signals for each protein from the western blots presented in Fig. 8e. Band intensity measured on ImageJ for each protein in each sample was divided by the corresponding intensity of Akt signal (used as the loading control).

Source data

Supplementary information

Source data

Source Data Fig. 1

Unprocessed immunoblot of Fig. 1d.

Source Data Fig. 1

Statistical source data for Fig. 1.

Source Data Fig. 2

Unprocessed immunoblot of Fig. 2f.

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. 8

Statistical source data.

Source Data Fig. 8

Unprocessed immunoblot of Fig. 8.

Source Data Extended Data Fig. 1

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. 6

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 10

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Nedeva, C., Menassa, J., Duan, M. et al. TREML4 receptor regulates inflammation and innate immune cell death during polymicrobial sepsis. Nat Immunol 21, 1585–1596 (2020). https://doi.org/10.1038/s41590-020-0789-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-020-0789-z

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