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:

DEL-1 promotes macrophage efferocytosis and clearance of inflammation

Abstract

Resolution of inflammation is essential for tissue homeostasis and represents a promising approach to inflammatory disorders. Here we found that developmental endothelial locus-1 (DEL-1), a secreted protein that inhibits leukocyte–endothelial adhesion and inflammation initiation, also functions as a non-redundant downstream effector in inflammation clearance. In human and mouse periodontitis, waning of inflammation was correlated with DEL-1 upregulation, whereas resolution of experimental periodontitis failed in DEL-1 deficiency. This concept was mechanistically substantiated in acute monosodium-urate-crystal-induced inflammation, where the pro-resolution function of DEL-1 was attributed to effective apoptotic neutrophil clearance (efferocytosis). DEL-1-mediated efferocytosis induced liver X receptor–dependent macrophage reprogramming to a pro-resolving phenotype and was required for optimal production of at least certain specific pro-resolving mediators. Experiments in transgenic mice with cell-specific overexpression of DEL-1 linked its anti-leukocyte-recruitment action to endothelial cell–derived DEL-1 and its efferocytic/pro-resolving action to macrophage-derived DEL-1. Thus, the compartmentalized expression of DEL-1 facilitates distinct homeostatic functions in an appropriate context that can be harnessed therapeutically.

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: DEL-1 concentration is elevated in the GCF following resolution of periodontitis due to treatment with SRP.
Fig. 2: The pro-resolution effect of DEL-1 in a resolving model of mouse LIP.
Fig. 3: DEL-1 modulates resolution of acute inflammation.
Fig. 4: DEL-1 enhances efferocytic activity through interaction with phosphatidylserine and αvβ3 integrin.
Fig. 5: DEL-1 promotes phenotypic alterations in macrophages during inflammation resolution through LXR.
Fig. 6: Compartmentalized expression of DEL-1 regulates resolution of acute inflammation.

Similar content being viewed by others

Data availability:

The data that support the findings of this study are available from the corresponding authors upon reasonable request.

References

  1. Fullerton, J. N. & Gilroy, D. W. Resolution of inflammation: a new therapeutic frontier. Nat. Rev. Drug. Discov. 15, 551–567 (2016).

    Article  CAS  PubMed  Google Scholar 

  2. Serhan, C. N. Pro-resolving lipid mediators are leads for resolution physiology. Nature 510, 92–101 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Serhan, C. N., Chiang, N. & Dalli, J. The resolution code of acute inflammation: novel pro-resolving lipid mediators in resolution. Semin. Immunol. 27, 200–215 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Greenlee-Wacker, M. C. Clearance of apoptotic neutrophils and resolution of inflammation. Immunol. Rev. 273, 357–370 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Birge, R. B. et al. Phosphatidylserine is a global immunosuppressive signal in efferocytosis, infectious disease, and cancer. Cell Death Differ. 23, 962–978 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Ortega-Gómez, A., Perretti, M. & Soehnlein, O. Resolution of inflammation: an integrated view. EMBO Mol. Med. 5, 661–674 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  7. A-Gonzalez, N. & Hidalgo, A. Nuclear receptors and clearance of apoptotic cells: stimulating the macrophage’s appetite. Front. Immunol. 5, 211 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Hajishengallis, G. & Chavakis, T. Endogenous modulators of inflammatory cell recruitment. Trends Immunol. 34, 1–6 (2013).

    Article  CAS  PubMed  Google Scholar 

  9. Shin, J. et al. DEL-1 restrains osteoclastogenesis and inhibits inflammatory bone loss in nonhuman primates. Sci Transl. Med. 7, 307ra155 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  10. Mitroulis, I. et al. Secreted protein Del-1 regulates myelopoiesis in the hematopoietic stem cell niche. J. Clin. Investig. 127, 3624–3639 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Choi, E. Y. et al. Del-1, an endogenous leukocyte-endothelial adhesion inhibitor, limits inflammatory cell recruitment. Science 322, 1101–1104 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Mitroulis, I. et al. Developmental endothelial locus-1 attenuates complement-dependent phagocytosis through inhibition of Mac-1-integrin. Thromb. Haemost. 111, 1004–1006 (2014).

    Article  CAS  PubMed  Google Scholar 

  13. Choi, E. Y. et al. Developmental endothelial locus-1 is a homeostatic factor in the central nervous system limiting neuroinflammation and demyelination. Mol. Psychiatry 20, 880–888 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Eskan, M. A. et al. The leukocyte integrin antagonist Del-1 inhibits IL-17-mediated inflammatory bone loss. Nat. Immunol. 13, 465–473 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Kang, Y.-Y., Kim, D.-Y., Lee, S.-H. & Choi, E. Y. Deficiency of developmental endothelial locus-1 (Del-1) aggravates bleomycin-induced pulmonary fibrosis in mice. Biochem. Biophys. Res. Commun. 445, 369–374 (2014).

    Article  CAS  PubMed  Google Scholar 

  16. Kourtzelis, I. et al. Developmental endothelial locus-1 modulates platelet-monocyte interactions and instant blood-mediated inflammatory reaction in islet transplantation. Thromb. Haemost. 115, 781–788 (2016).

    Article  PubMed  Google Scholar 

  17. Maekawa, T. et al. Antagonistic effects of IL-17 and D-resolvins on endothelial Del-1 expression through a GSK-3β-C/EBPβ pathway. Nat. Commun. 6, 8272 (2015).

    Article  PubMed  Google Scholar 

  18. Folwaczny, M., Karnesi, E., Berger, T. & Paschos, E. Clinical association between chronic periodontitis and the leukocyte extravasation inhibitors developmental endothelial locus-1 and pentraxin-3. Eur. J. Oral Sci. 125, 258–264 (2017).

    Article  CAS  PubMed  Google Scholar 

  19. Hajishengallis, G., Moutsopoulos, N. M., Hajishengallis, E. & Chavakis, T. Immune and regulatory functions of neutrophils in inflammatory bone loss. Semin. Immunol. 28, 146–158 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Kantarci, A. & Van Dyke, T. E. Resolution of inflammation in periodontitis. J. Periodontol. 76, 2168–2174 (2005).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Kajikawa, T. et al. Milk fat globule epidermal growth factor 8 inhibits periodontitis in non-human primates and its gingival crevicular fluid levels can differentiate periodontal health from disease in humans. J. Clin. Periodontol. 44, 472–483 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Cohen, R. E., Research, Science and Therapy Committee, American Academy of Periodontology. Position paper: periodontal maintenance. J. Periodontol. 74, 1395–1401 (2003).

    Article  PubMed  Google Scholar 

  23. Hajishengallis, G., Lamont, R. J. & Graves, D. T. The enduring importance of animal models in understanding periodontal disease. Virulence 6, 229–235 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Ariel, A. & Timor, O. Hanging in the balance: endogenous anti-inflammatory mechanisms in tissue repair and fibrosis. J. Pathol. 229, 250–263 (2013).

    Article  CAS  PubMed  Google Scholar 

  25. Choi, E. Y. et al. Regulation of LFA-1-dependent inflammatory cell recruitment by Cbl-b and 14-3-3 proteins. Blood 111, 3607–3614 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Getting, S. J. et al. Molecular determinants of monosodium urate crystal-induced murine peritonitis: a role for endogenous mast cells and a distinct requirement for endothelial-derived selectins. J. Pharmacol. Exp. Ther. 283, 123–130 (1997).

    CAS  PubMed  Google Scholar 

  27. Martin, W. J., Shaw, O., Liu, X., Steiger, S. & Harper, J. L. Monosodium urate monohydrate crystal-recruited noninflammatory monocytes differentiate into M1-like proinflammatory macrophages in a peritoneal murine model of gout. Arthritis Rheum. 63, 1322–1332 (2011).

    Article  CAS  PubMed  Google Scholar 

  28. Serhan, C. N., Chiang, N. & Van Dyke, T. E. Resolving inflammation: dual anti-inflammatory and pro-resolution lipid mediators. Nat. Rev. Immunol. 8, 349–361 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Dalli, J. et al. Resolvin D3 and aspirin-triggered resolvin D3 are potent immunoresolvents. Chem. Biol. 20, 188–201 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Bannenberg, G. L. et al. Molecular circuits of resolution: formation and actions of resolvins and protectins. J. Immunol. 174, 4345–4355 (2005).

    Article  CAS  PubMed  Google Scholar 

  31. Hong, S. et al. Resolvin E1 metabolome in local inactivation during inflammation-resolution. J. Immunol. 180, 3512–3519 (2008).

    Article  CAS  PubMed  Google Scholar 

  32. Villoutreix, B. O. & Miteva, M. A. Discoidin domains as emerging therapeutic targets. Trends Pharmacol. Sci. 37, 641–659 (2016).

    Article  CAS  PubMed  Google Scholar 

  33. Lastrucci, C. et al. Molecular and cellular profiles of the resolution phase in a damage-associated molecular pattern (DAMP)-mediated peritonitis model and revelation of leukocyte persistence in peritoneal tissues. FASEB J. 29, 1914–1929 (2015).

    Article  CAS  PubMed  Google Scholar 

  34. Savill, J., Dransfield, I., Hogg, N. & Haslett, C. Vitronectin receptor-mediated phagocytosis of cells undergoing apoptosis. Nature 343, 170–173 (1990).

    Article  CAS  PubMed  Google Scholar 

  35. Mevorach, D., Mascarenhas, J. O., Gershov, D. & Elkon, K. B. Complement-dependent clearance of apoptotic cells by human macrophages. J. Exp. Med. 188, 2313–2320 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Stables, M. J. et al. Transcriptomic analyses of murine resolution-phase macrophages. Blood 118, e192–e208 (2011).

    Article  CAS  PubMed  Google Scholar 

  37. Rébé, C. et al. Induction of transglutaminase 2 by a liver X receptor/retinoic acid receptor αpathway increases the clearance of apoptotic cells by human macrophages. Circ. Res. 105, 393–401 (2009).

    Article  PubMed  Google Scholar 

  38. Hanayama, R., Tanaka, M., Miwa, K. & Nagata, S. Expression of developmental endothelial locus-1 in a subset of macrophages for engulfment of apoptotic cells. J. Immunol. 172, 3876–3882 (2004).

    Article  CAS  PubMed  Google Scholar 

  39. Rosas, M. et al. The transcription factor Gata6 links tissue macrophage phenotype and proliferative renewal. Science 344, 645–648 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Hajishengallis, G. Periodontitis: from microbial immune subversion to systemic inflammation. Nat. Rev. Immunol. 15, 30–44 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Chiang, N. et al. Infection regulates pro-resolving mediators that lower antibiotic requirements. Nature 484, 524–528 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Hasturk, H. et al. Resolvin E1 regulates inflammation at the cellular and tissue level and restores tissue homeostasis in vivo. J. Immunol. 179, 7021–7029 (2007).

    Article  CAS  PubMed  Google Scholar 

  43. A-Gonzalez, N. et al. Apoptotic cells promote their own clearance and immune tolerance through activation of the nuclear receptor LXR. Immunity 31, 245–258 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Kidani, Y. & Bensinger, S. J. Liver X receptor and peroxisome proliferator-activated receptor as integrators of lipid homeostasis and immunity. Immunol. Rev. 249, 72–83 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Choi, J.-Y. et al. Mer signaling increases the abundance of the transcription factor LXR to promote the resolution of acute sterile inflammation. Sci. Signal. 8, ra21 (2015).

    Article  CAS  PubMed  Google Scholar 

  46. Hong, C. et al. Constitutive activation of LXR in macrophages regulates metabolic and inflammatory gene expression: identification of ARL7 as a direct target. J. Lipid Res. 52, 531–539 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Hidai, C., Kawana, M., Kitano, H. & Kokubun, S. Discoidin domain of Del1 protein contributes to its deposition in the extracellular matrix. Cell Tissue Res. 330, 83–95 (2007).

    Article  CAS  PubMed  Google Scholar 

  48. Galli, S. J., Borregaard, N. & Wynn, T. A. Phenotypic and functional plasticity of cells of innate immunity: macrophages, mast cells and neutrophils. Nat. Immunol. 12, 1035–1044 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Matzinger, P. & Kamala, T. Tissue-based class control: the other side of tolerance. Nat. Rev. Immunol. 11, 221–230 (2011).

    Article  CAS  PubMed  Google Scholar 

  50. Griffiths, G. S. Formation, collection and significance of gingival crevice fluid. Periodontol. 2000 31, 32–42 (2003).

    Article  PubMed  Google Scholar 

  51. Bostanci, N. et al. Gingival crevicular fluid levels of RANKL and OPG in periodontal diseases: implications of their relative ratio. J. Clin. Periodontol. 34, 370–376 (2007).

    Article  CAS  PubMed  Google Scholar 

  52. Subramanian, P. et al. Endothelial cell-specific overexpression of developmental endothelial locus-1 does not influence atherosclerosis development in ApoE−/− mice. Thromb. Haemost. 117, 2003–2005 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  53. Chen, L. S. et al. Endothelial cell-specific overexpression of Del-1 drives expansion of haematopoietic progenitor cells in the bone marrow. Thromb. Haemost. 118, 613–616 (2018).

  54. Lang, R., Rutschman, R. L., Greaves, D. R. & Murray, P. J. Autocrine deactivation of macrophages in transgenic mice constitutively overexpressing IL-10 under control of the human CD68 promoter. J. Immunol. 168, 3402–3411 (2002).

    Article  CAS  PubMed  Google Scholar 

  55. Gough, P. J., Gordon, S. & Greaves, D. R. The use of human CD68 transcriptional regulatory sequences to direct high-level expression of class A scavenger receptor in macrophages in vitro and in vivo. Immunology 103, 351–361 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Chung, K.-J. et al. A self-sustained loop of inflammation-driven inhibition of beige adipogenesis in obesity. Nat. Immunol. 18, 654–664 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Coxon, A. et al. A novel role for the β2 integrin CD11b/CD18 in neutrophil apoptosis: a homeostatic mechanism in inflammation. Immunity 5, 653–666 (1996).

    Article  PubMed  Google Scholar 

  58. A-Gonzalez, N. et al. The nuclear receptor LXRα controls the functional specialization of splenic macrophages. Nat. Immunol. 14, 831–839 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Joosten, L. A. B. et al. Engagement of fatty acids with Toll-like receptor 2 drives interleukin-1β production via the ASC/caspase 1 pathway in monosodium urate monohydrate crystal-induced gouty arthritis. Arthritis Rheum. 62, 3237–3248 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Graves, D. T., Fine, D., Teng, Y.-T. A., Van Dyke, T. E. & Hajishengallis, G. The use of rodent models to investigate host-bacteria interactions related to periodontal diseases. J. Clin. Periodontol. 35, 89–105 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  61. Abe, T. & Hajishengallis, G. Optimization of the ligature-induced periodontitis model in mice. J. Immunol. Methods 394, 49–54 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Matsuda, S. et al. A novel method of sampling gingival crevicular fluid from a mouse model of periodontitis. J. Immunol. Methods 438, 21–25 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Abe, T. et al. The B cell-stimulatory cytokines BLyS and APRIL are elevated in human periodontitis and are required for B cell-dependent bone loss in experimental murine periodontitis. J. Immunol. 195, 1427–1435 (2015).

    Article  CAS  PubMed  Google Scholar 

  64. Mukundan, L. et al. PPAR-δ senses and orchestrates clearance of apoptotic cells to promote tolerance. Nat. Med. 15, 1266–1272 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Xiong, W., Frasch, S. C., Thomas, S. M., Bratton, D. L. & Henson, P. M. Induction of TGF-β1 synthesis by macrophages in response to apoptotic cells requires activation of the scavenger receptor CD36. PLoS ONE 8, e72772 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Fadok, V. A. et al. Macrophages that have ingested apoptotic cells in vitro inhibit proinflammatory cytokine production through autocrine/paracrine mechanisms involving TGF-β, PGE2, and PAF. J. Clin. Investig. 101, 890–898 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Mitroulis, I. et al. Modulation of myelopoiesis progenitors is an integral component of trained immunity. Cell 172, 147–161 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Davies, L. C. et al. A quantifiable proliferative burst of tissue macrophages restores homeostatic macrophage populations after acute inflammation. Eur. J. Immunol. 41, 2155–2164 (2011).

    Article  CAS  PubMed  Google Scholar 

  69. Gautier, E. L. et al. Gata6 regulates aspartoacylase expression in resident peritoneal macrophages and controls their survival. J. Exp. Med. 211, 1525–1531 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by grants from the European Research Council (DEMETINL to T.C.), the Deutsche Forschungsgemeinschaft (Transregio-SFB 83 to Ü.C. as well as Transregio-SFB 127 and Transregio-SFB 205 to T.C.), and the National Institutes of Health (AI068730, DE024153, DE024716, DE026152, and DE015254 to G.H.). I.K. was supported by the MeDDriveGrant (60.400) from the Technische Universität Dresden Medical Faculty. A.Castrillo was supported by Spanish MINECO SAF2014-56819R and SAF2015-71878-REDT. This work was supported in part by intramural National Institute of Dental and Craniofacial Research, National Institutes of Health funding. We thank R. Naumann (Transgenic Core Facility, Max-Planck Institute for Cell Biology and Genetics Dresden, Germany) for microinjections and B. Gercken and M. Schuster (Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany) for technical assistance. Parts of this work are included in the master’s theses of C.D. at the Technische Universität Dresden, Dresden, Germany, and A.F. at the University of Porto, Porto, Portugal.

Author information

Authors and Affiliations

Authors

Contributions

I.K. designed the study, performed experiments, analyzed and interpreted data, and wrote the manuscript; X.L. and I.M. designed and performed experiments and analyzed and interpreted data; D.G. generated reagents and performed experiments; T.K. performed experiments and analyzed and interpreted data; B.Wang and J.M.K. designed experiments and interpreted data; M.G., J.v.R., A.Czogalla, A.F., and C.D. performed experiments and interpreted data; M.T., K.R., L.-S.C., and K.-J.C. performed experiments; K.H. and S.G. generated critical reagents and performed experiments; J.-H.L. and A.K.T. interpreted data; L.A.B.J., N.M.M, B.Wielockx, and A.Castrillo contributed to experimental design and interpreted data; Ü.C. supervised research, designed experiments, and interpreted data; and G.H. and T.C. conceived and designed the study, supervised research, interpreted data, and wrote the paper.

Corresponding authors

Correspondence to George Hajishengallis or Triantafyllos Chavakis.

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.

Integrated supplementary information

Supplementary Figure 1 The role of DEL-1 in resolution of inflammation in periodontitis.

a, Probing pocket depth (PPD) was determined in human patients diagnosed with periodontitis before and after scaling and root planing (SRP). b, The protein concentration of DEL-1 was measured in the supernatant of peritoneal exudates obtained from wild-type (WT) or Del-1KO mice at steady state by ELISA (pg/mg of total protein; n = 4 mice per group). c, Relative mRNA expression of the indicated molecules in gingival tissue from wild-type (WT) or Del1KO mice, which were subjected to LIP for 5 d and were euthanized immediately (5 days ligated; 5DL) or 5 d later with the ligatures removed to enable resolution (5 days ligated and 5 days with ligatures removed; 5DL-5DR). Data were normalized to Gapdh mRNA and are presented as fold change relative to baseline (NL, not ligated), which was set as 1 for each genotype (n = 5 mice per group). d, Wild-type (WT) or Del1KO mice were subjected to LIP for up to 5 or 10 d and ligatures were removed or not at day 5 (NL, not ligated; 5DL, 5 days ligated; 10DL, 10 days ligated; 5DL-5DR, 5 days ligated and 5 days with ligatures removed). Numbers of neutrophils in gingival tissues were assessed by immunostaining (n = 6 mice per group). eg, Wild-type (WT) and Del1KO mice were subjected to LIP and ligatures were removed on day 5. RvD1 (100 nM) or PBS control was then administered on a daily basis until the day before sacrifice, which was performed on day 10 (that is, total of five doses). e, Bone loss was measured on day 10 (n = 6 mice per group). f,g, The concentrations of IL-17A and TGF-β1 in gingival tissue were assayed by ELISA on day 10 (n = 6 mice per group; pg of cytokines/mg of total protein are shown). a, P < 0.0001, pre-post paired two-way ANOVA with repeated measures by patient. bg, *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001, NS, non-significant. b,c, Two-tailed Mann–Whitney U test; d, two-way ANOVA with Sidak’s multiple-comparisons test; e, one-way ANOVA, Sidak’s multiple-comparisons test; f,g, two-tailed Student’s t test. Data are means +/− s.e.m. and are derived from one experiment (b,c) or pooled from two experiments (dg).

Supplementary Figure 2 Peritoneal leukocyte analysis in MSU crystal–induced inflammation.

a, Representative FACS plot from a wild-type mouse at 24 h after intraperitoneal MSU crystal injection showing the gating strategy of leukocytes present in peritoneal exudates. R1, neutrophils; R2, monocytes/macrophages; R3, eosinophils. b, Numbers of monocytes/macrophages (Mono+Mac) in peritoneal exudates obtained from wild-type (WT) and Del1KO mice 8 h (n = 11 WT and 13 Del1KO mice) and 24 h (n = 18 WT and 16 Del1KO mice) after MSU crystal–induced inflammation. c,d, Numbers of neutrophils (c) and monocytes/macrophages (Mono+Mac) (d) in peritoneal exudates obtained from wild-type (WT) mice and mice deficient in αL integrin (ItgalKO) 8 h after MSU crystal–induced inflammation (n = 6 WT and 7 ItgalKO mice). e,f, Wild-type mice were injected intravenously with 50 μg of a neutralizing antibody against αM integrin (anti-αM) or isotype control and then were treated with MSU crystals. The numbers of peritoneal neutrophils (e) and monocytes/macrophages (Mono+Mac) (f) were assessed 8 h after MSU injection (n = 6 mice per group). g, Relative mRNA expression of Gpr18, Chemr23 and Fpr2 in peritoneal monocytes/macrophages sorted from wild-type (WT) and Del1KO mice 24 h after MSU-induced inflammation. Relative mRNA expression was normalized against 18S and was set as 1 in wild-type monocytes/macrophages (n = 5 WT and 4 Del1KO mice). h, Wild-type (WT) and Del1KO mice were treated with MSU crystals and RvD1 or control PBS (–). The relative number of apoptotic neutrophils in peritoneal exudates 24 h after treatment is shown. Numbers of apoptotic neutrophils are expressed relative to the WT control (–) group set as 1 (n = 13 mice for the WT and WT+RvD1 groups and n = 12 mice for the Del1KO and Del1KO+RvD1 groups). *P < 0.05, **P < 0.01. NS, non-significant. b, Two-tailed Student’s t test; cg, two-tailed Mann–Whitney U test; h, one-way ANOVA with Tukey’s multiple-comparisons test. Data are means +/− s.e.m. and are pooled from two experiments (b, 8 h time point and h), from three experiments (b, 24 h time point) or are derived from one experiment (cg).

Supplementary Figure 3 The effect of DEL-1 on macrophage gene expression at steady state or in the setting of efferocytosis.

(a), Relative mRNA expression of Tgfb1 and Lxra in peritoneal F4/80+CD11b+ macrophages sorted from untreated wild-type (WT) or Del1KO mice (not subjected to MSU crystal–induced inflammation). Relative expression was normalized against 18S and was set as 1 in WT macrophages (n = 6 WT and 9 Del1KO mice). (b,c), Relative mRNA expression of Tgfb1 in wild-type bone marrow-derived macrophages (BMDMs) (n = 6 separate cell isolations) (b) and isolated peritoneal macrophages from untreated wild-type mice (n = 8 separate cell isolations) (c) upon co-culture of either with mouse apoptotic neutrophils for 3 h in the presence of Fc control or DEL-1 Fc (each 500 ng/ml). (di), Relative mRNA expression of Lxra (n = 11 separate cell isolations) (d), Lxrb (n = 12 separate cell isolations) (e), Ucp2 (n =12 separate cell isolations) (f), Abca1 (n = 12 separate cell isolations) (g), Tgm2 (n = 6 separate cell isolations) (h), and Rxra (n = 6 separate cell isolations) (i) in wild-type BMDMs upon co-culture with mouse apoptotic neutrophils for 3 h in the presence of Fc control or DEL-1 Fc (each 500 ng/ml). (j), Relative mRNA expression of Tgfb1 and Lxra in wild-type BMDMs treated with Fc control or DEL-1 Fc (500 ng/ml each; n = 6 separate cell isolations for Tgfb1 and 4 separate cell isolations for Lxra). In (bj), relative expression was set as 1 in cells that were treated with Fc control in each case. Relative mRNA expression was normalized against 18S. *P < 0.05, **P < 0.01. NS, non-significant. (a,b,e,f,h–j), Two-tailed Mann–Whitney U test; (c,d,g), two-tailed Student’s t test. Data are means +/− s.e.m. and are derived from one experiment (a), from one experiment representative of two (b,hj), or are pooled from two experiments (cg).

Supplementary Figure 4 The impact of specific sites of DEL-1 and of LXR signaling on the phenotype of efferocytic macrophages.

(ac), Relative mRNA expression of Tgfb1 (a), Lxra (b), and Lxrb (c) in wild-type bone marrow-derived macrophages (BMDMs) upon co-culture with apoptotic neutrophils for 3 h in the presence of Fc control, DEL-1 that is mutated in the RGD motif (DEL-1[RGE] Fc) or the truncated protein, in which discoidin I–like domains are absent (DEL-1[E1-E3] Fc) (each 500 ng/ml) is shown (n = 6 separate cell isolations in (a,b) and 12 separate cell isolations in c). d, Shown is relative Lxra and Tgfb1 mRNA expression in Itgb3KO BMDMs that were co-cultured with apoptotic neutrophils for 3 h in the presence of Fc control or DEL-1 Fc (each 500 ng/ml) (n = 5 separate cell isolations). In (ad), gene expression was normalized against 18S; relative expression was set as 1 in BMDMs that were treated with Fc in each case. (e), Relative Tgfb1 mRNA expression in wild-type BMDMs that were treated with DEL-1 Fc and co-cultured with mouse apoptotic neutrophils for 3 h in the presence of an LXR antagonist (1 μM) or vehicle control (DMSO; Ctrl). Relative mRNA expression was normalized against 18S; relative expression was set as 1 in the presence of vehicle control (n = 15 separate cell isolations). (f,g), Relative Tgfb1 mRNA expression in wild-type BMDMs treated with an LXR antagonist (1 μM) or vehicle control (DMSO; Ctrl) (n = 16 separate cell isolations) (f) and untreated BMDMs from wild-type (WT) mice or from mice deficient in both LXRα and LXRβ (Lxra-LxrbDKO) (n = 5 separate cell isolations per genotype) (g). Relative expression was normalized against 18S and was set as 1 in BMDMs treated with vehicle control (Ctrl) (f) or in wild-type BMDMs (g). *P < 0.05. NS, non-significant. (ac), One-way ANOVA with Dunnett’s multiple-comparisons test; (d,f,g), two-tailed Mann–Whitney U test; e, two-tailed Student’s t test. Data are means +/− s.e.m. and are from one experiment representative of two experiments (a,b), pooled from two (c) or three (e,f) experiments, or derived from one experiment (d,g).

Supplementary Figure 5 Characterization of mice overexpressing DEL-1 or segments thereof in macrophages.

(a,b), Relative mRNA expression of Del1 in isolated peritoneal macrophages and bone marrow-derived macrophages (BMDMs) from untreated (not subjected to MSU crystal–induced inflammation) mice that overexpress in macrophages full-length DEL-1 (CD68-Del1 mice) (a) or the truncated version of DEL-1 that lacks the discoidin I–like domains (CD68-Del1-[E1-E3] mice) (b) and from their respective untreated wild-type (WT) mice. Relative expression was normalized against 18S and was set as 1 in macrophages from WT mice in each case (a, n = 6 WT and 8 CD68-Del1 mice; (b), n = 6 mice per group). (c,d), Numbers of neutrophils (c) and relative number of apoptotic neutrophils (d) in peritoneal exudates obtained from CD68-Del1-[E1-E3] mice and wild-type (WT) mice 24 h after MSU crystal–induced inflammation. Numbers of apoptotic neutrophils are expressed relative to the WT group, set as 1 (n = 8 mice per group). **P < 0.01, NS, non-significant. (a,b,d), Two-tailed Mann–Whitney U test; c, two-tailed Student’s t test. Data are means +/− s.e.m. and are pooled from two (a) or three (bd) experiments.

Supplementary Figure 6 The role of integrins on DEL-1-dependent efferocytosis and liposome quality control.

(a,b), Isolated peritoneal macrophages from untreated wild-type (WT) or CD68-Del1 mice were co-cultured for 30 min with apoptotic neutrophils that were prestained with BCECF-AM in the presence of IgG2b (a) or IgG2a (b) control antibody, neutralizing antibody against Mac-1 integrin or against ICAM-1 (a), or antibody against LFA-1 integrin (b) (each antibody 10 μg/ml). Relative efferocytosis index is shown and was calculated as the ratio of macrophages that have phagocytosed apoptotic material to the total number of macrophages. Data are expressed relative to the control group (a, WT-IgG2b; b, WT-IgG2a), set as 1 (n = 5 separate cell isolations per genotype). (c), Representative TLC of liposomes composed of PC/Chol. or PC/Chol./PS stained with primuline. Lanes 1–3 show lipid standards; PC (lane 1), PS (lane 2), cholesterol (Chol., lane 3). (d), Zeta potential measurements of PC/Chol. and PC/Chol./PS liposomes are presented (n = 3 separate preparations). **P < 0.01, NS, non-significant. (a,b), One-way ANOVA with Tukey’s multiple-comparisons test. Data are means +/− s.e.m. and are from three experiments (a,b,d). The experiment in c is representative of three.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–6 and Supplementary Table 1

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kourtzelis, I., Li, X., Mitroulis, I. et al. DEL-1 promotes macrophage efferocytosis and clearance of inflammation. Nat Immunol 20, 40–49 (2019). https://doi.org/10.1038/s41590-018-0249-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-018-0249-1

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