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.

  • Review Article
  • Published:

How macrophages deal with death

Abstract

Tissue macrophages rapidly recognize and engulf apoptotic cells. These events require the display of so-called eat-me signals on the apoptotic cell surface, the most fundamental of which is phosphatidylserine (PtdSer). Externalization of this phospholipid is catalysed by scramblase enzymes, several of which are activated by caspase cleavage. PtdSer is detected both by macrophage receptors that bind to this phospholipid directly and by receptors that bind to a soluble bridging protein that is independently bound to PtdSer. Prominent among the latter receptors are the MER and AXL receptor tyrosine kinases. Eat-me signals also trigger macrophages to engulf virus-infected or metabolically traumatized, but still living, cells, and this ‘murder by phagocytosis’ may be a common phenomenon. Finally, the localized presentation of PtdSer and other eat-me signals on delimited cell surface domains may enable the phagocytic pruning of these ‘locally dead’ domains by macrophages, most notably by microglia of the central nervous system.

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: Receptors and ligands mediating apoptotic cell recognition and phagocytosis by macrophages.
Fig. 2: Phosphatidylserine-triggered phagocytic engulfment of living cells.
Fig. 3: Phosphatidylserine-delimited phagocytosis of only small parts of cells.

Similar content being viewed by others

References

  1. Nagata, S. Apoptosis and clearance of apoptotic cells. Annu. Rev. Immunol. 36, 489–517 (2018).

    CAS  PubMed  Google Scholar 

  2. Elliott, M. R. & Ravichandran, K. S. The dynamics of apoptotic cell clearance. Dev. Cell 38, 147–160 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Gordon, S. & Pluddemann, A. Macrophage clearance of apoptotic cells: a critical assessment. Front. Immunol. 9, 127 (2018).

    PubMed  PubMed Central  Google Scholar 

  4. Epelman, S., Lavine, K. J. & Randolph, G. J. Origin and functions of tissue macrophages. Immunity 41, 21–35 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Krysko, D. V., Vanden Berghe, T., D’Herde, K. & Vandenabeele, P. Apoptosis and necrosis: detection, discrimination and phagocytosis. Methods 44, 205–221 (2008).

    CAS  PubMed  Google Scholar 

  6. deCathelineau, A. M. & Henson, P. M. The final step in programmed cell death: phagocytes carry apoptotic cells to the grave. Essays Biochem. 39, 105–117 (2003).

    CAS  PubMed  Google Scholar 

  7. Surh, C. D. & Sprent, J. T cell apoptosis detected in situ during positive and negative selection in the thymus. Nature 372, 100–103 (1994).

    CAS  PubMed  Google Scholar 

  8. Earnshaw, W. C., Martins, L. M. & Kaufmann, S. H. Mammalian caspases: structure, activation, substrates, and functions during apoptosis. Annu. Rev. Biochem. 68, 383–424 (1999).

    CAS  PubMed  Google Scholar 

  9. Mahajan, A., Herrmann, M. & Munoz, L. E. Clearance deficiency and cell death pathways: a model for the pathogenesis of SLE. Front. Immunol. 7, 35 (2016).

    PubMed  PubMed Central  Google Scholar 

  10. Abdolmaleki, F. et al. The role of efferocytosis in autoimmune diseases. Front. Immunol. 9, 1645 (2018).

    PubMed  PubMed Central  Google Scholar 

  11. Fourgeaud, L. et al. TAM receptors regulate multiple features of microglial physiology. Nature 532, 240–244 (2016). This paper highlights a role for microglial MER in the steady state phagocytosis of apoptotic cells in the neurogenic regions of the adult brain.

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Sierra, A. et al. Microglia shape adult hippocampal neurogenesis through apoptosis-coupled phagocytosis. Cell Stem Cell 7, 483–495 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. McGaha, T. L. & Karlsson, M. C. Apoptotic cell responses in the splenic marginal zone: a paradigm for immunologic reactions to apoptotic antigens with implications for autoimmunity. Immunol. Rev. 269, 26–43 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Peter, C., Wesselborg, S., Herrmann, M. & Lauber, K. Dangerous attraction: phagocyte recruitment and danger signals of apoptotic and necrotic cells. Apoptosis 15, 1007–1028 (2010).

    PubMed  Google Scholar 

  15. Medina, C. B. & Ravichandran, K. S. Do not let death do us part: ‘find-me’ signals in communication between dying cells and the phagocytes. Cell Death Differ. 23, 979–989 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Lauber, K. et al. Apoptotic cells induce migration of phagocytes via caspase-3-mediated release of a lipid attraction signal. Cell 113, 717–730 (2003).

    CAS  PubMed  Google Scholar 

  17. Peter, C. et al. Migration to apoptotic “find-me” signals is mediated via the phagocyte receptor G2A. J. Biol. Chem. 283, 5296–5305 (2008).

    CAS  PubMed  Google Scholar 

  18. Gude, D. R. et al. Apoptosis induces expression of sphingosine kinase 1 to release sphingosine-1-phosphate as a “come-and-get-me” signal. FASEB J. 22, 2629–2638 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Elliott, M. R. et al. Nucleotides released by apoptotic cells act as a find-me signal to promote phagocytic clearance. Nature 461, 282–286 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Peter, C. et al. Release of lysophospholipid ‘find-me’ signals during apoptosis requires the ATP-binding cassette transporter A1. Autoimmunity 45, 568–573 (2012).

    CAS  PubMed  Google Scholar 

  21. Wu, Y. C. & Horvitz, H. R. The C. elegans cell corpse engulfment gene ced-7 encodes a protein similar to ABC transporters. Cell 93, 951–960 (1998).

    CAS  PubMed  Google Scholar 

  22. Chekeni, F. B. et al. Pannexin 1 channels mediate ‘find-me’ signal release and membrane permeability during apoptosis. Nature 467, 863–867 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Sandilos, J. K. et al. Pannexin 1, an ATP release channel, is activated by caspase cleavage of its pore-associated C-terminal autoinhibitory region. J. Biol. Chem. 287, 11303–11311 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Sandilos, J. K. & Bayliss, D. A. Physiological mechanisms for the modulation of pannexin 1 channel activity. J. Physiol. 590, 6257–6266 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Chiu, Y. H. et al. A quantized mechanism for activation of pannexin channels. Nat. Commun. 8, 14324 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Truman, L. A. et al. CX3CL1/fractalkine is released from apoptotic lymphocytes to stimulate macrophage chemotaxis. Blood 112, 5026–5036 (2008).

    CAS  PubMed  Google Scholar 

  27. Yang, L. V., Radu, C. G., Wang, L., Riedinger, M. & Witte, O. N. Gi-independent macrophage chemotaxis to lysophosphatidylcholine via the immunoregulatory GPCR G2A. Blood 105, 1127–1134 (2005).

    CAS  PubMed  Google Scholar 

  28. Luo, B. et al. Erythropoeitin signaling in macrophages promotes dying cell clearance and immune tolerance. Immunity 44, 287–302 (2016).

    CAS  PubMed  Google Scholar 

  29. Idzko, M., Ferrari, D. & Eltzschig, H. K. Nucleotide signalling during inflammation. Nature 509, 310–317 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Haynes, S. E. et al. The P2Y12 receptor regulates microglial activation by extracellular nucleotides. Nat. Neurosci. 9, 1512–1519 (2006).

    CAS  PubMed  Google Scholar 

  31. Gautier, E. L. et al. Gene-expression profiles and transcriptional regulatory pathways that underlie the identity and diversity of mouse tissue macrophages. Nat. Immunol. 13, 1118–1128 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. ImmGen, C. Open-source ImmGen: mononuclear phagocytes. Nat. Immunol. 17, 741 (2016).

    Google Scholar 

  33. Ohsawa, K. et al. P2Y12 receptor-mediated integrin-beta1 activation regulates microglial process extension induced by ATP. Glia 58, 790–801 (2010).

    PubMed  Google Scholar 

  34. Zrzavy, T. et al. Loss of ‘homeostatic’ microglia and patterns of their activation in active multiple sclerosis. Brain 140, 1900–1913 (2017).

    PubMed  PubMed Central  Google Scholar 

  35. Kronlage, M. et al. Autocrine purinergic receptor signaling is essential for macrophage chemotaxis. Sci. Signal. 3, ra55 (2010).

    PubMed  Google Scholar 

  36. Fadok, V. A. et al. Exposure of phosphatidylserine on the surface of apoptotic lymphocytes triggers specific recognition and removal by macrophages. J. Immunol. 148, 2207–2216 (1992). This paper identifies externalized PtdSer as an eat-me signal for phagocytosis of apoptotic cells.

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Leventis, P. A. & Grinstein, S. The distribution and function of phosphatidylserine in cellular membranes. Annu. Rev. Biophys. 39, 407–427 (2010).

    CAS  PubMed  Google Scholar 

  39. van Meer, G. Dynamic transbilayer lipid asymmetry. Cold Spring Harb. Perspect. Biol. 3, a004671 (2011).

    PubMed  PubMed Central  Google Scholar 

  40. Andersen, J. P. et al. P4-ATPases as phospholipid flippases-structure, function, and enigmas. Front. Physiol. 7, 275 (2016).

    PubMed  PubMed Central  Google Scholar 

  41. Shin, H. W. & Takatsu, H. Substrates of P4-ATPases: beyond aminophospholipids (phosphatidylserine and phosphatidylethanolamine). FASEB J. 33, 3087–3096 (2018).

    PubMed  Google Scholar 

  42. Nagata, S., Suzuki, J., Segawa, K. & Fujii, T. Exposure of phosphatidylserine on the cell surface. Cell Death Differ. 23, 952–961 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Pomorski, T. G. & Menon, A. K. Lipid somersaults: uncovering the mechanisms of protein-mediated lipid flipping. Prog. Lipid Res. 64, 69–84 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Tang, X., Halleck, M. S., Schlegel, R. A. & Williamson, P. A subfamily of P-type ATPases with aminophospholipid transporting activity. Science 272, 1495–1497 (1996).

    CAS  PubMed  Google Scholar 

  45. Segawa, K., Kurata, S. & Nagata, S. Human type IV P-type ATPases that work as plasma membrane phospholipid flippases and their regulation by caspase and calcium. J. Biol. Chem. 291, 762–772 (2016).

    CAS  PubMed  Google Scholar 

  46. Segawa, K. et al. Caspase-mediated cleavage of phospholipid flippase for apoptotic phosphatidylserine exposure. Science 344, 1164–1168 (2014). This paper identifies ATP11C as a PtdSer flippase whose enzymatic activity is lost upon caspase cleavage.

    CAS  PubMed  Google Scholar 

  47. Segawa, K. et al. Phospholipid flippases enable precursor B cells to flee engulfment by macrophages. Proc. Natl Acad. Sci. USA 115, 12212–12217 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Coleman, J. A. & Molday, R. S. Critical role of the beta-subunit CDC50A in the stable expression, assembly, subcellular localization, and lipid transport activity of the P4-ATPase ATP8A2. J. Biol. Chem. 286, 17205–17216 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Kornberg, R. D. & McConnell, H. M. Inside-outside transitions of phospholipids in vesicle membranes. Biochemistry 10, 1111–1120 (1971).

    CAS  PubMed  Google Scholar 

  50. Whitlock, J. M. & Hartzell, H. C. Anoctamins/TMEM16 proteins: chloride channels flirting with lipids and extracellular vesicles. Annu. Rev. Physiol. 79, 119–143 (2017).

    CAS  PubMed  Google Scholar 

  51. Suzuki, J. et al. Calcium-dependent phospholipid scramblase activity of TMEM16 protein family members. J. Biol. Chem. 288, 13305–13316 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Suzuki, J., Imanishi, E. & Nagata, S. Xkr8 phospholipid scrambling complex in apoptotic phosphatidylserine exposure. Proc. Natl Acad. Sci. USA 113, 9509–9514 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Suzuki, J., Umeda, M., Sims, P. J. & Nagata, S. Calcium-dependent phospholipid scrambling by TMEM16F. Nature 468, 834–838 (2010). This paper identifies a widely expressed PtdSer scramblase that is activated by Ca 2+ binding.

    CAS  PubMed  Google Scholar 

  54. Suzuki, J., Denning, D. P., Imanishi, E., Horvitz, H. R. & Nagata, S. Xk-related protein 8 and CED-8 promote phosphatidylserine exposure in apoptotic cells. Science 341, 403–406 (2013). This paper identifies a widely expressed PtdSer scramblase that is activated by caspase cleavage.

    CAS  PubMed  Google Scholar 

  55. Suzuki, J., Imanishi, E. & Nagata, S. Exposure of phosphatidylserine by Xk-related protein family members during apoptosis. J. Biol. Chem. 289, 30257–30267 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Bevers, E. M. & Williamson, P. L. Getting to the outer leaflet: physiology of phosphatidylserine exposure at the plasma membrane. Physiol. Rev. 96, 605–645 (2016).

    CAS  PubMed  Google Scholar 

  57. Yu, K. et al. Identification of a lipid scrambling domain in ANO6/TMEM16F. eLife 4, e06901 (2015).

    PubMed  PubMed Central  Google Scholar 

  58. Watanabe, R., Sakuragi, T., Noji, H. & Nagata, S. Single-molecule analysis of phospholipid scrambling by TMEM16F. Proc. Natl Acad. Sci. USA 115, 3066–3071 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Fujii, T., Sakata, A., Nishimura, S., Eto, K. & Nagata, S. TMEM16F is required for phosphatidylserine exposure and microparticle release in activated mouse platelets. Proc. Natl Acad. Sci. USA 112, 12800–12805 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Freeman, G. J., Casasnovas, J. M., Umetsu, D. T. & DeKruyff, R. H. TIM genes: a family of cell surface phosphatidylserine receptors that regulate innate and adaptive immunity. Immunol. Rev. 235, 172–189 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Lemke, G. Biology of the TAM receptors. Cold Spring Harb. Perspect. Biol. 5, a009076 (2013).

    PubMed  PubMed Central  Google Scholar 

  62. Miyanishi, M. et al. Identification of Tim4 as a phosphatidylserine receptor. Nature 450, 435–439 (2007). This paper describes TIM4 as a direct PtdSer binder.

    CAS  PubMed  Google Scholar 

  63. Yanagihashi, Y., Segawa, K., Maeda, R., Nabeshima, Y. I. & Nagata, S. Mouse macrophages show different requirements for phosphatidylserine receptor Tim4 in efferocytosis. Proc. Natl Acad. Sci. USA 114, 8800–8805 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Flannagan, R. S., Canton, J., Furuya, W., Glogauer, M. & Grinstein, S. The phosphatidylserine receptor TIM4 utilizes integrins as coreceptors to effect phagocytosis. Mol. Biol. Cell 25, 1511–1522 (2014).

    PubMed  PubMed Central  Google Scholar 

  65. Wong, K. et al. Phosphatidylserine receptor Tim-4 is essential for the maintenance of the homeostatic state of resident peritoneal macrophages. Proc. Natl Acad. Sci. USA 107, 8712–8717 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Rodriguez-Manzanet, R. et al. T and B cell hyperactivity and autoimmunity associated with niche-specific defects in apoptotic body clearance in TIM-4-deficient mice. Proc. Natl Acad. Sci. USA 107, 8706–8711 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Santiago, C. et al. Structures of T cell immunoglobulin mucin protein 4 show a metal-Ion-dependent ligand binding site where phosphatidylserine binds. Immunity 27, 941–951 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. DeKruyff, R. H. et al. T cell/transmembrane, Ig, and mucin-3 allelic variants differentially recognize phosphatidylserine and mediate phagocytosis of apoptotic cells. J. Immunol. 184, 1918–1930 (2010).

    CAS  PubMed  Google Scholar 

  69. Tietjen, G. T. et al. Molecular mechanism for differential recognition of membrane phosphatidylserine by the immune regulatory receptor Tim4. Proc. Natl Acad. Sci. USA 111, E1463–E1472 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Lemke, G. & Rothlin, C. V. Immunobiology of the TAM receptors. Nat. Rev. Immunol. 8, 327–336 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Lew, E. D. et al. Differential TAM receptor-ligand-phospholipid interactions delimit differential TAM bioactivities. eLife 3, e03385 (2014). This paper establishes the rules of engagement for TAM receptors and their ligands.

    PubMed Central  Google Scholar 

  72. Stitt, T. N. et al. The anticoagulation factor protein S and its relative, Gas6, are ligands for the Tyro 3/Axl family of receptor tyrosine kinases. Cell 80, 661–670 (1995).

    CAS  PubMed  Google Scholar 

  73. Dransfield, I., Zagorska, A., Lew, E. D., Michail, K. & Lemke, G. Mer receptor tyrosine kinase mediates both tethering and phagocytosis of apoptotic cells. Cell Death Dis. 6, e1646 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Zagórska, A., Través, P. G., Lew, E. D., Dransfield, I. & Lemke, G. Diversification of TAM receptor tyrosine kinase function. Nat. Immunol. 15, 920–928 (2014). This paper identifies MER and AXL as mediators of efferocytosis under homeostatic and inflammatory conditions, respectively.

    PubMed  PubMed Central  Google Scholar 

  75. Lemke, G. Phosphatidylserine is the signal for TAM receptors and their ligands. Trends Biochem. Sci. 42, 738–748 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Rothlin, C. V., Carrera-Silva, E. A., Bosurgi, L. & Ghosh, S. TAM receptor signaling in immune homeostasis. Annu. Rev. Immunol. 33, 355–391 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Fujimori, T. et al. The Axl receptor tyrosine kinase is a discriminator of macrophage function in the inflamed lung. Mucosal Immunol. 8, 1021–1030 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Tsou, W. I. et al. Receptor tyrosine kinases, TYRO3, AXL and MER, demonstrate distinct patterns and complex regulation of ligand-induced activation. J. Biol. Chem. 289, 25750–25763 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Scott, R. S. et al. Phagocytosis and clearance of apoptotic cells is mediated by MER. Nature 411, 207–211 (2001). This paper provides the first evidence for MER as a mediator of efferocytosis.

    CAS  PubMed  Google Scholar 

  80. Grabiec, A. M., Goenka, A., Fife, M. E., Fujimori, T. & Hussell, T. Axl and MerTK receptor tyrosine kinases maintain human macrophage efferocytic capacity in the presence of viral triggers. Eur. J. Immunol. 48, 855–860 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Cohen, P. L. et al. Delayed apoptotic cell clearance and lupus-like autoimmunity in mice lacking the c-mer membrane tyrosine kinase. J. Exp. Med. 196, 135–140 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Thorp, E., Cui, D., Schrijvers, D. M., Kuriakose, G. & Tabas, I. Mertk receptor mutation reduces efferocytosis efficiency and promotes apoptotic cell accumulation and plaque necrosis in atherosclerotic lesions of apoe−/− mice. Arterioscler. Thromb. Vasc. Biol. 28, 1421–1428 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Toda, S., Segawa, K. & Nagata, S. MerTK-mediated engulfment of pyrenocytes by central macrophages in erythroblastic islands. Blood 123, 3963–3971 (2014).

    CAS  PubMed  Google Scholar 

  85. Ancot, F., Foveau, B., Lefebvre, J., Leroy, C. & Tulasne, D. Proteolytic cleavages give receptor tyrosine kinases the gift of ubiquity. Oncogene 28, 2185–2195 (2009).

    CAS  PubMed  Google Scholar 

  86. Orme, J. J. et al. Heightened cleavage of Axl receptor tyrosine kinase by ADAM metalloproteases may contribute to disease pathogenesis in SLE. Clin. Immunol. 169, 58–68 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  87. Dengler, M. et al. Accurate determination of soluble Axl by enzyme-linked immunosorbent assay. Assay Drug Dev. Technol. 14, 543–550 (2016).

    CAS  PubMed  Google Scholar 

  88. Rothlin, C. V., Ghosh, S., Zuniga, E. I., Oldstone, M. B. & Lemke, G. TAM receptors are pleiotropic inhibitors of the innate immune response. Cell 131, 1124–1136 (2007).This paper delineates a mechanism whereby AXL functions as an intrinsic negative feedback inhibitor of the innate immune response in DCs.

    CAS  PubMed  Google Scholar 

  89. Chan, P. Y. et al. The TAM family receptor tyrosine kinase TYRO3 is a negative regulator of type 2 immunity. Science 352, 99–103 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Lu, Q. et al. Tyro-3 family receptors are essential regulators of mammalian spermatogenesis. Nature 398, 723–728 (1999).

    CAS  PubMed  Google Scholar 

  91. Lu, Q. & Lemke, G. Homeostatic regulation of the immune system by receptor tyrosine kinases of the Tyro 3 family. Science 293, 306–311 (2001).This paper describes TAM receptor expression in macrophages and the autoimmune phenotypes that appear in mice with mutations in TAM receptor genes.

    CAS  PubMed  Google Scholar 

  92. Khan, T. N., Wong, E. B., Soni, C. & Rahman, Z. S. Prolonged apoptotic cell accumulation in germinal centers of Mer-deficient mice causes elevated B cell and CD4+ Th cell responses leading to autoantibody production. J. Immunol. 190, 1433–1446 (2013).

    CAS  PubMed  Google Scholar 

  93. Lemke, G. & Lu, Q. Macrophage regulation by Tyro 3 family receptors. Curr. Opin. Immunol. 15, 31–36 (2003).

    CAS  PubMed  Google Scholar 

  94. Schell, S. L. et al. Mer receptor tyrosine kinase signaling prevents self-ligand sensing and aberrant selection in germinal centers. J. Immunol. 199, 4001–4015 (2017).

    CAS  PubMed  Google Scholar 

  95. Wallet, M. A. et al. MerTK is required for apoptotic cell-induced T cell tolerance. J. Exp. Med. 205, 219–232 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Kaminska, A., Enguita, F. J. & Stepien, E. L. Lactadherin: an unappreciated haemostasis regulator and potential therapeutic agent. Vascul. Pharmacol. 101, 21–28 (2018).

    CAS  PubMed  Google Scholar 

  97. Hanayama, R. et al. Identification of a factor that links apoptotic cells to phagocytes. Nature 417, 182–187 (2002). This paper identifies MFGE8 as a soluble bridging protein that binds to PtdSer.

    CAS  PubMed  Google Scholar 

  98. Hanayama, R. et al. Autoimmune disease and impaired uptake of apoptotic cells in MFG-E8-deficient mice. Science 304, 1147–1150 (2004).

    CAS  PubMed  Google Scholar 

  99. Akakura, S. et al. The opsonin MFG-E8 is a ligand for the alphavbeta5 integrin and triggers DOCK180-dependent Rac1 activation for the phagocytosis of apoptotic cells. Exp. Cell Res. 292, 403–416 (2004).

    CAS  PubMed  Google Scholar 

  100. Raymond, A., Ensslin, M. A. & Shur, B. D. SED1/MFG-E8: a bi-motif protein that orchestrates diverse cellular interactions. J. Cell. Biochem. 106, 957–966 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Peng, Y. & Elkon, K. B. Autoimmunity in MFG-E8-deficient mice is associated with altered trafficking and enhanced cross-presentation of apoptotic cell antigens. J. Clin. Invest. 121, 2221–2241 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Lauber, K. et al. Milk fat globule-EGF factor 8 mediates the enhancement of apoptotic cell clearance by glucocorticoids. Cell Death Differ. 20, 1230–1240 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Murakami, Y. et al. CD300b regulates the phagocytosis of apoptotic cells via phosphatidylserine recognition. Cell Death Differ. 21, 1746–1757 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  104. Borrego, F. The CD300 molecules: an emerging family of regulators of the immune system. Blood 121, 1951–1960 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Park, D. et al. BAI1 is an engulfment receptor for apoptotic cells upstream of the ELMO/Dock180/Rac module. Nature 450, 430–434 (2007).

    CAS  PubMed  Google Scholar 

  106. Das, S. et al. Brain angiogenesis inhibitor 1 (BAI1) is a pattern recognition receptor that mediates macrophage binding and engulfment of Gram-negative bacteria. Proc. Natl Acad. Sci. USA 108, 2136–2141 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  107. Zhu, D. et al. BAI1 regulates spatial learning and synaptic plasticity in the hippocampus. J. Clin. Invest. 125, 1497–1508 (2015).

    PubMed  PubMed Central  Google Scholar 

  108. Lee, C. S. et al. Boosting apoptotic cell clearance by colonic epithelial cells attenuates inflammation in vivo. Immunity 44, 807–820 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Park, S. Y. et al. Rapid cell corpse clearance by stabilin-2, a membrane phosphatidylserine receptor. Cell Death Differ. 15, 192–201 (2008).

    CAS  PubMed  Google Scholar 

  110. Kim, S. et al. Cross talk between engulfment receptors stabilin-2 and integrin alphavbeta5 orchestrates engulfment of phosphatidylserine-exposed erythrocytes. Mol. Cell. Biol. 32, 2698–2708 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  111. Taylor, P. R. et al. A hierarchical role for classical pathway complement proteins in the clearance of apoptotic cells in vivo. J. Exp. Med. 192, 359–366 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  112. Ramirez-Ortiz, Z. G. et al. The scavenger receptor SCARF1 mediates the clearance of apoptotic cells and prevents autoimmunity. Nat. Immunol. 14, 917–926 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  113. Galvan, M. D., Greenlee-Wacker, M. C. & Bohlson, S. S. C1q and phagocytosis: the perfect complement to a good meal. J. Leukoc. Biol. 92, 489–497 (2012).

    CAS  PubMed  Google Scholar 

  114. Martin, M. & Blom, A. M. Complement in removal of the dead — balancing inflammation. Immunol. Rev. 274, 218–232 (2016).

    CAS  PubMed  Google Scholar 

  115. Paidassi, H. et al. C1q binds phosphatidylserine and likely acts as a multiligand-bridging molecule in apoptotic cell recognition. J. Immunol. 180, 2329–2338 (2008).

    CAS  PubMed  Google Scholar 

  116. Stegert, M., Bock, M. & Trendelenburg, M. Clinical presentation of human C1q deficiency: how much of a lupus? Mol. Immunol. 67, 3–11 (2015).

    CAS  PubMed  Google Scholar 

  117. Clarke, E. V., Weist, B. M., Walsh, C. M. & Tenner, A. J. Complement protein C1q bound to apoptotic cells suppresses human macrophage and dendritic cell-mediated Th17 and Th1 T cell subset proliferation. J. Leukoc. Biol. 97, 147–160 (2015).

    PubMed  Google Scholar 

  118. Kenyon, K. D. et al. IgG autoantibodies against deposited C3 inhibit macrophage-mediated apoptotic cell engulfment in systemic autoimmunity. J. Immunol. 187, 2101–2111 (2011).

    CAS  PubMed  Google Scholar 

  119. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  120. Suh, C. H., Hilliard, B., Li, S., Merrill, J. T. & Cohen, P. L. TAM receptor ligands in lupus: protein S but not Gas6 levels reflect disease activity in systemic lupus erythematosus. Arthritis Res. Ther. 12, R146 (2010).

    PubMed  PubMed Central  Google Scholar 

  121. Gao, A. G. et al. Integrin-associated protein is a receptor for the C-terminal domain of thrombospondin. J. Biol. Chem. 271, 21–24 (1996).

    CAS  PubMed  Google Scholar 

  122. Brown, E. J. & Frazier, W. A. Integrin-associated protein (CD47) and its ligands. Trends Cell Biol. 11, 130–135 (2001).

    CAS  PubMed  Google Scholar 

  123. Oldenborg, P. A. et al. Role of CD47 as a marker of self on red blood cells. Science 288, 2051–2054 (2000).

    CAS  PubMed  Google Scholar 

  124. Barclay, A. N. & Van den Berg, T. K. The interaction between signal regulatory protein alpha (SIRPalpha) and CD47: structure, function, and therapeutic target. Annu. Rev. Immunol. 32, 25–50 (2014).

    CAS  PubMed  Google Scholar 

  125. Bian, Z. et al. Cd47-Sirpalpha interaction and IL-10 constrain inflammation-induced macrophage phagocytosis of healthy self-cells. Proc. Natl Acad. Sci. USA 113, E5434–E5443 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  126. Jaiswal, S. et al. CD47 is upregulated on circulating hematopoietic stem cells and leukemia cells to avoid phagocytosis. Cell 138, 271–285 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  127. Chao, M. P. et al. Anti-CD47 antibody synergizes with rituximab to promote phagocytosis and eradicate non-Hodgkin lymphoma. Cell 142, 699–713 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  128. Horrigan, S. K. & Reproducibility Project: Cancer Biology. Replication study: the CD47-signal regulatory protein alpha (SIRPa) interaction is a therapeutic target for human solid tumors. eLife 6, e18173 (2017).

    PubMed  PubMed Central  Google Scholar 

  129. Russ, A. et al. Blocking “don’t eat me” signal of CD47-SIRPalpha in hematological malignancies, an in-depth review. Blood Rev. 32, 480–489 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  130. Lehrman, E. K. et al. CD47 protects synapses from excess microglia-mediated pruning during development. Neuron 100, 120–134 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  131. Gumienny, T. L. et al. CED-12/ELMO, a novel member of the CrkII/Dock180/Rac pathway, is required for phagocytosis and cell migration. Cell 107, 27–41 (2001).

    CAS  PubMed  Google Scholar 

  132. Tosello-Trampont, A. C., Brugnera, E. & Ravichandran, K. S. Evidence for a conserved role for CRKII and Rac in engulfment of apoptotic cells. J. Biol. Chem. 276, 13797–13802 (2001).

    CAS  PubMed  Google Scholar 

  133. Leverrier, Y. & Ridley, A. J. Requirement for Rho GTPases and PI 3-kinases during apoptotic cell phagocytosis by macrophages. Curr. Biol. 11, 195–199 (2001).

    CAS  PubMed  Google Scholar 

  134. Kitano, M., Nakaya, M., Nakamura, T., Nagata, S. & Matsuda, M. Imaging of Rab5 activity identifies essential regulators for phagosome maturation. Nature 453, 241–245 (2008).

    CAS  PubMed  Google Scholar 

  135. Fond, A. M. & Ravichandran, K. S. Clearance of dying cells by phagocytes: mechanisms and implications for disease pathogenesis. Adv. Exp. Med. Biol 930, 25–49 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  136. Park, S. Y. & Kim, I. S. Engulfment signals and the phagocytic machinery for apoptotic cell clearance. Exp. Mol. Med. 49, e331 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  137. Hochreiter-Hufford, A. & Ravichandran, K. S. Clearing the dead: apoptotic cell sensing, recognition, engulfment, and digestion. Cold Spring Harb. Perspect. Biol. 5, a008748 (2013).

    PubMed  PubMed Central  Google Scholar 

  138. Hoeppner, D. J., Hengartner, M. O. & Schnabel, R. Engulfment genes cooperate with ced-3 to promote cell death in Caenorhabditis elegans. Nature 412, 202–206 (2001).

    CAS  PubMed  Google Scholar 

  139. Reddien, P. W., Cameron, S. & Horvitz, H. R. Phagocytosis promotes programmed cell death in C. elegans. Nature 412, 198–202 (2001).

    CAS  PubMed  Google Scholar 

  140. Tufail, Y. et al. Phosphatidylserine exposure controls viral innate immune responses by microglia. Neuron 93, 574–586 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  141. Neher, J. J. et al. Phagocytosis executes delayed neuronal death after focal brain ischemia. Proc. Natl Acad. Sci. USA 110, E4098–E4107 (2013). This paper provides in vivo evidence that live neurons may be phagocytosed by microglia in a MER-dependent process.

    CAS  PubMed  PubMed Central  Google Scholar 

  142. Brelstaff, J., Tolkovsky, A. M., Ghetti, B., Goedert, M. & Spillantini, M. G. Living neurons with Tau filaments aberrantly expose phosphatidylserine and are phagocytosed by microglia. Cell Rep. 24, 1939–1948 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  143. Fricker, M. et al. MFG-E8 mediates primary phagocytosis of viable neurons during neuroinflammation. J. Neurosci. 32, 2657–2666 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  144. Chua, B. A. et al. Protein S and Gas6 induce efferocytosis of HIV-1-infected cells. Virology 515, 176–190 (2018).

    CAS  PubMed  Google Scholar 

  145. Brown, G. C. & Neher, J. J. Microglial phagocytosis of live neurons. Nat. Rev. Neurosci. 15, 209–216 (2014).

    CAS  PubMed  Google Scholar 

  146. Fricker, M., Tolkovsky, A. M., Borutaite, V., Coleman, M. & Brown, G. C. Neuronal cell death. Physiol. Rev. 98, 813–880 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  147. Strauss, O. The retinal pigment epithelium in visual function. Physiol. Rev. 85, 845–881 (2005).

    CAS  PubMed  Google Scholar 

  148. Sparrow, J. R., Hicks, D. & Hamel, C. P. The retinal pigment epithelium in health and disease. Curr. Mol. Med. 10, 802–823 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  149. D’Cruz, P. M. et al. Mutation of the receptor tyrosine kinase gene Mertk in the retinal dystrophic RCS rat. Hum. Mol. Genet. 9, 645–651 (2000).

    PubMed  Google Scholar 

  150. Duncan, J. L. et al. An RCS-like retinal dystrophy phenotype in mer knockout mice. Invest. Ophthalmol. Vis. Sci. 44, 826–838 (2003).

    PubMed  Google Scholar 

  151. Gal, A. et al. Mutations in MERTK, the human orthologue of the RCS rat retinal dystrophy gene, cause retinitis pigmentosa. Nat. Genet. 26, 270–271 (2000). This paper describes the first of a series of human MERTK mutations that result in retinal dystrophies.

    CAS  PubMed  Google Scholar 

  152. Audo, I. et al. MERTK mutation update in inherited retinal diseases. Hum. Mutat. 39, 887–913 (2018).

    CAS  PubMed  Google Scholar 

  153. Ghazi, N. G. et al. Treatment of retinitis pigmentosa due to MERTK mutations by ocular subretinal injection of adeno-associated virus gene vector: results of a phase I trial. Hum. Genet. 135, 327–343 (2016).

    CAS  PubMed  Google Scholar 

  154. Burstyn-Cohen, T. et al. Genetic dissection of TAM receptor-ligand interaction in retinal pigment epithelial cell phagocytosis. Neuron 76, 1123–1132 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  155. Nandrot, E. F. & Finnemann, S. C. Lack of alphavbeta5 integrin receptor or its ligand MFG-E8: distinct effects on retinal function. Ophthalm. Res. 40, 120–123 (2008).

    CAS  Google Scholar 

  156. Ruggiero, L., Connor, M. P., Chen, J., Langen, R. & Finnemann, S. C. Diurnal, localized exposure of phosphatidylserine by rod outer segment tips in wild-type but not Itgb5−/− or Mfge8−/− mouse retina. Proc. Natl Acad. Sci. USA 109, 8145–8148 (2012). This paper presents evidence that localized PtdSer externalization specifies the localized phagocytic excision of photoreceptor outer segments.

    CAS  PubMed  PubMed Central  Google Scholar 

  157. Hong, S., Dissing-Olesen, L. & Stevens, B. New insights on the role of microglia in synaptic pruning in health and disease. Curr. Opin. Neurobiol. 36, 128–134 (2016).

    CAS  PubMed  Google Scholar 

  158. Thion, M. S. & Garel, S. Microglia under the spotlight: activity and complement-dependent engulfment of synapses. Trends Neurosci. 41, 332–334 (2018).

    CAS  PubMed  Google Scholar 

  159. Schafer, D. P. et al. Microglia sculpt postnatal neural circuits in an activity and complement-dependent manner. Neuron 74, 691–705 (2012). This paper describes a role for complement decoration of presynaptic neuronal elements that are phagocytosed by microglia.

    CAS  PubMed  PubMed Central  Google Scholar 

  160. Presumey, J., Bialas, A. R. & Carroll, M. C. Complement system in neural synapse elimination in development and disease. Adv. Immunol. 135, 53–79 (2017).

    CAS  PubMed  Google Scholar 

  161. Sekar, A. et al. Schizophrenia risk from complex variation of complement component 4. Nature 530, 177–183 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  162. Gyorffy, B. A. et al. Local apoptotic-like mechanisms underlie complement-mediated synaptic pruning. Proc. Natl Acad. Sci. USA 115, 6303–6308 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  163. Bhattacharyya, S. et al. Enveloped viruses disable innate immune responses in dendritic cells by direct activation of TAM receptors. Cell Host Microbe 14, 136–147 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  164. Moller-Tank, S. & Maury, W. Phosphatidylserine receptors: enhancers of enveloped virus entry and infection. Virology 468–470, 565–580 (2014).

    PubMed  Google Scholar 

  165. Schrijvers, D. M., De Meyer, G. R., Kockx, M. M., Herman, A. G. & Martinet, W. Phagocytosis of apoptotic cells by macrophages is impaired in atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 25, 1256–1261 (2005).

    CAS  PubMed  Google Scholar 

  166. Yurdagul, A. et al. Mechanisms and consequences of defective efferocytosis in atherosclerosis. Front. Cardiovasc. Med. 4, 86 (2017).

    PubMed  Google Scholar 

  167. Grabiec, A. M. & Hussell, T. The role of airway macrophages in apoptotic cell clearance following acute and chronic lung inflammation. Semin. Immunopathol. 38, 409–423 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  168. Baumann, I. et al. Impaired uptake of apoptotic cells into tingible body macrophages in germinal centers of patients with systemic lupus erythematosus. Arthritis Rheum. 46, 191–201 (2002).

    PubMed  Google Scholar 

  169. Schoumacher, M. & Burbridge, M. Key roles of AXL and MER receptor tyrosine kinases in resistance to multiple anticancer therapies. Curr. Oncol. Rep. 19, 19 (2017).

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The author thanks the members of his laboratory for comments and discussions. This work is supported by grants from the US National Institutes of Health (NIH) (AI101400, NS085296 and AG060748), the Cure Alzheimer’s Fund and the Leona M. and Harry B. Helmsley Charitable Trust.

Reviewer information

Nature Reviews Immunology thanks S. Grinstein and the other anonymous reviewer(s) for their contribution to the peer review of this work.

Author information

Authors and Affiliations

Authors

Contributions

G.L. was responsible for researching data for this article, for the discussion of content and for the writing, review and editing of the manuscript before submission.

Corresponding author

Correspondence to Greg Lemke.

Ethics declarations

Competing interests

The author declares no competing interests.

Additional information

Publisher’s note

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

Glossary

NIH3T3 cells

A cell line derived from mouse embryonic fibroblasts.

AD293 cells

A cell line derived from the HEK293 human embryonic kidney cell line.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lemke, G. How macrophages deal with death. Nat Rev Immunol 19, 539–549 (2019). https://doi.org/10.1038/s41577-019-0167-y

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41577-019-0167-y

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