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:

Emerging connectivity of programmed cell death pathways and its physiological implications

Abstract

The removal of functionally dispensable, infected or potentially neoplastic cells is driven by programmed cell death (PCD) pathways, highlighting their important roles in homeostasis, host defence against pathogens, cancer and a range of other pathologies. Several types of PCD pathways have been described, including apoptosis, necroptosis and pyroptosis; they employ distinct molecular and cellular processes and differ in their outcomes, such as the capacity to trigger inflammatory responses. Recent genetic and biochemical studies have revealed remarkable flexibility in the use of these PCD pathways and indicate a considerable degree of plasticity in their molecular regulation; for example, despite having a primary role in inducing pyroptosis, inflammatory caspases can also induce apoptosis, and conversely, apoptotic stimuli can trigger pyroptosis. Intriguingly, this flexibility is most pronounced in cellular responses to infection, while apoptosis is the dominant cell death process through which organisms prevent the development of cancer. In this Review, we summarize the mechanisms of the different types of PCD and describe the physiological and pathological processes that engage crosstalk between these pathways, focusing on infections and cancer. We discuss the intriguing notion that the different types of PCD could be seen as a single, coordinated cell death system, in which the individual pathways are highly interconnected and can flexibly compensate for one another.

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: Different forms of programmed cell death lead to different bystander responses.
Fig. 2: Molecular mechanisms of apoptosis pathway activation.
Fig. 3: Molecular features of inflammasome activation and pyroptosis.
Fig. 4: Induction of necroptosis.
Fig. 5: The role of cell death in host responses to infection.
Fig. 6: The molecular mechanisms of cell survival regulation by TNFR1 signalling.
Fig. 7: Overlap and backup of apoptosis and pyroptosis as means to induce cell death.

Similar content being viewed by others

References

  1. Thompson, C. B. Apoptosis in the pathogenesis and treatment of disease. Science 267, 1456–1462 (1995).

    CAS  PubMed  Google Scholar 

  2. Ellis, R. E., Yuan, J. & Horvitz, H. R. Mechanisms and functions of cell death. Annu. Rev. Cell Biol. 7, 663–698 (1991).

    CAS  PubMed  Google Scholar 

  3. Strasser, A., O’Connor, L. & Dixit, V. M. Apoptosis signaling. Ann. Rev. Biochem. 69, 217–245 (2000).

    CAS  PubMed  Google Scholar 

  4. Green, D. R. The coming decade of cell death research: five riddles. Cell 177, 1094–1107 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Ke, F. F. S. et al. Embryogenesis and adult life in the absence of intrinsic apoptosis Effectors BAX, BAK, and BOK. Cell 173, 1217–1230 e1217 (2018).

    CAS  PubMed  Google Scholar 

  6. Lindsten, T. et al. The combined functions of proapoptotic Bcl-2 family members Bak and Bax are essential for normal development of multiple tissues. Mol. Cell 6, 1389–1399 (2000). This report describes the essential overlapping role of BAX and BAK for the execution of the intrinsic pathway of apoptosis during embryogenesis and tissue homeostasis.

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Murphy, J. M. et al. The pseudokinase MLKL mediates necroptosis via a molecular switch mechanism. Immunity 39, 443–453 (2013).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  9. Sun, L. et al. Mixed lineage kinase domain-like protein mediates necrosis signaling downstream of RIP3 kinase. Cell 148, 213–227 (2012). Together with Murphy et al. (2013), this study identifies MLKL as the essential executioner of necroptosis.

    CAS  PubMed  Google Scholar 

  10. Rock, K. L. & Kono, H. The inflammatory response to cell death. Annu. Rev. Pathol. 3, 99–126 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Van Opdenbosch, N. & Lamkanfi, M. Caspases in cell death, inflammation, and disease. Immunity 50, 1352–1364 (2019).

    PubMed  PubMed Central  Google Scholar 

  12. Boatright, K. M. & Salvesen, G. S. Mechanisms of caspase activation. Curr. Opin. Cell Biol. 15, 725–731 (2003).

    CAS  PubMed  Google Scholar 

  13. Kerr, J. F. R., Wyllie, A. H. & Currie, A. R. Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics. Brit. J. Cancer 26, 239–257 (1972). This publication coins the term ‘apoptotis’ and describes the morphology observed during an apoptotic process.

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Segawa, K. et al. Caspase-mediated cleavage of phospholipid flippase for apoptotic phosphatidylserine exposure. Science 344, 1164–1168 (2014).

    CAS  PubMed  Google Scholar 

  15. Czabotar, P. E., Lessene, G., Strasser, A. & Adams, J. M. Control of apoptosis by the BCL-2 protein family: implications for physiology and therapy. Nat. Rev. Mol. Cell Biol. 15, 49–63 (2014).

    CAS  PubMed  Google Scholar 

  16. Singh, R., Letai, A. & Sarosiek, K. Regulation of apoptosis in health and disease: the balancing act of BCL-2 family proteins. Nat. Rev. Mol. Cell Biol. 20, 175–193 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Cosentino, K. & Garcia-Saez, A. J. Bax and Bak pores: Are we closing the circle? Trends Cell Biol. 27, 266–275 (2017).

    CAS  PubMed  Google Scholar 

  18. O’Neill, K. L., Huang, K., Zhang, J., Chen, Y. & Luo, X. Inactivation of prosurvival Bcl-2 proteins activates Bax/Bak through the outer mitochondrial membrane. Genes Dev. 30, 973–988 (2016).

    PubMed  PubMed Central  Google Scholar 

  19. Salvesen, G. S. & Dixit, V. M. Caspase activation: the induced-proximity model. Proc. Natl Acad. Sci. USA 96, 10964–10967 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Galban, S. & Duckett, C. S. XIAP as a ubiquitin ligase in cellular signaling. Cell Death Differ. 17, 54–60 (2010).

    CAS  PubMed  Google Scholar 

  21. Ekert, P. G. & Vaux, D. L. The mitochondrial death squad: hardened killers or innocent bystanders? Curr. Opin. Cell Biol. 17, 626–630 (2005).

    CAS  PubMed  Google Scholar 

  22. Ashkenazi, A. & Dixit, V. M. Death receptors: signaling and modulation. Science 281, 1305–1308 (1998).

    CAS  PubMed  Google Scholar 

  23. Kischkel, F. C. et al. Cytotoxicity-dependent APO-1 (Fas/CD95)-associated proteins form a death-inducing signaling complex (DISC) with the receptor. EMBO J. 14, 5579–5588 (1995). Kischkel et al. provide a description of the death-inducing signalling complex that is critical for FAS to induce apoptosis.

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Kataoka, T. The caspase-8 modulator c-FLIP. Crit. Rev. Immunol. 25, 31–58 (2005).

    CAS  PubMed  Google Scholar 

  25. Billen, L. P., Shamas-Din, A. & Andrews, D. W. Bid: a Bax-like BH3 protein. Oncogene 27, S93–S104 (2008).

    CAS  PubMed  Google Scholar 

  26. Brennan, M. A. & Cookson, B. T. Salmonella induces macrophage death by caspase-1-dependent necrosis. Mol. Microbiol. 38, 31–40 (2000).

    CAS  PubMed  Google Scholar 

  27. Sansonetti, P. J. et al. Caspase-1 activation of IL-1β and IL-18 are essential for Shigella flexneri-induced inflammation. Immunity 12, 581–590 (2000).

    CAS  PubMed  Google Scholar 

  28. Lamkanfi, M. & Dixit, V. M. Inflammasomes: guardians of cytosolic sanctity. Immunol. Rev. 227, 95–105 (2009).

    CAS  PubMed  Google Scholar 

  29. Swanson, K. V., Deng, M. & Ting, J. P. The NLRP3 inflammasome: molecular activation and regulation to therapeutics. Nat. Rev. Immunol. 19, 477–489 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Hornung, V. & Latz, E. Critical functions of priming and lysosomal damage for NLRP3 activation. Eur. J. Immunol. 40, 620–623 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. He, Y., Zeng, M. Y., Yang, D., Motro, B. & Nunez, G. NEK7 is an essential mediator of NLRP3 activation downstream of potassium efflux. Nature 530, 354–357 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Fernandes-Alnemri, T., Yu, J. W., Datta, P., Wu, J. & Alnemri, E. S. AIM2 activates the inflammasome and cell death in response to cytoplasmic DNA. Nature 458, 509–513 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Duncan, J. A. & Canna, S. W. The NLRC4 inflammasome. Immunol. Rev. 281, 115–123 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Broz, P., Pelegrin, P. & Shao, F. The gasdermins, a protein family executing cell death and inflammation. Nat. Rev. Immunol. 20, 143–157 (2020).

    CAS  PubMed  Google Scholar 

  35. Shi, J. et al. Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nature 526, 660–665 (2015). Together with Kayagaki et al. (2015), this study identifies GSDMD as the essential executioner of pyroptosis.

    CAS  PubMed  Google Scholar 

  36. Man, S. M., Kanneganti, T. D. & Gasdermin, D. the long-awaited executioner of pyroptosis. Cell Res. 25, 1183–1184 (2015).

    PubMed  PubMed Central  Google Scholar 

  37. Bergsbaken, T., Fink, S. L. & Cookson, B. T. Pyroptosis: host cell death and inflammation. Nat. Rev. Microbiol. 7, 99–109 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Kupz, A. et al. NLRC4 inflammasomes in dendritic cells regulate noncognate effector function by memory CD8+ T cells. Nat. Immunol. 13, 162–169 (2012).

    CAS  PubMed  Google Scholar 

  39. Ruhl, S. et al. ESCRT-dependent membrane repair negatively regulates pyroptosis downstream of GSDMD activation. Science 362, 956–960 (2018). Ruhl et al. demonstrate that the ESCRT machinery can protect cells from gasdermin-driven lysis at the early stages of pyroptosis.

    PubMed  Google Scholar 

  40. Salvamoser, R. et al. Characterisation of mice lacking the inflammatory caspases-1/11/12 reveals no contribution of caspase-12 to cell death and sepsis. Cell Death Differ. 26, 1124–1137 (2019).

    CAS  PubMed  Google Scholar 

  41. Wang, S. et al. Identification and characterization of Ich-3, a member of the interleukin-1β converting enzyme (ICE)/Ced-3 family and an upstream regulator of ICE. J. Biol. Chem. 271, 20580–20587 (1996).

    CAS  PubMed  Google Scholar 

  42. Kobori, M. et al. Wedelolactone suppresses LPS-induced caspase-11 expression by directly inhibiting the IKK complex. Cell Death Differ. 11, 123–130 (2004).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  44. Wang, S. et al. Murine caspase-11, an ICE-interacting protease, is essential for the activation of ICE. Cell 92, 501–509 (1998).

    CAS  PubMed  Google Scholar 

  45. Kayagaki, N. et al. Non-canonical inflammasome activation targets caspase-11. Nature 479, 117–122 (2011).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Lawlor, K. E. et al. XIAP loss triggers RIPK3- and caspase-8-driven IL-1β activation and cell death as a consequence of TLR-MyD88-induced cIAP1-TRAF2 degradation. Cell Rep. 20, 668–682 (2017).

    CAS  PubMed  Google Scholar 

  48. Lawlor, K. E. et al. RIPK3 promotes cell death and NLRP3 inflammasome activation in the absence of MLKL. Nat. Commun. 6, 6282 (2015).

    CAS  PubMed  Google Scholar 

  49. Newton, K. & Manning, G. Necroptosis and inflammation. Annu. Rev. Biochem. 85, 743–763 (2016).

    CAS  PubMed  Google Scholar 

  50. Newton, K. R. I. P. K. 1 and RIPK3: critical regulators of inflammation and cell death. Trends Cell Biol. 25, 347–353 (2015).

    CAS  PubMed  Google Scholar 

  51. Khan, N., Lawlor, K. E., Murphy, J. M. & Vince, J. E. More to life than death: molecular determinants of necroptotic and non-necroptotic RIP3 kinase signaling. Curr. Opin. Immunol. 26, 76–89 (2014).

    CAS  PubMed  Google Scholar 

  52. Gong, Y. N. et al. ESCRT-III acts downstream of MLKL to regulate necroptotic cell death and its consequences. Cell 169, 286–300 e216 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Vaux, D. L., Haecker, G. & Strasser, A. An evolutionary perspective on apoptosis. Cell 76, 777–779 (1994).

    CAS  PubMed  Google Scholar 

  54. Mariathasan, S. et al. Differential activation of the inflammasome by caspase-1 adaptors ASC and Ipaf. Nature 430, 213–218 (2004).

    CAS  PubMed  Google Scholar 

  55. Franchi, L., Eigenbrod, T., Munoz-Planillo, R. & Nunez, G. The inflammasome: a caspase-1-activation platform that regulates immune responses and disease pathogenesis. Nat. Immunol. 10, 241–247 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Sutterwala, F. S. et al. Immune recognition of Pseudomonas aeruginosa mediated by the IPAF/NLRC4 inflammasome. J. Exp. Med. 204, 3235–3245 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Fernandes-Alnemri, T. et al. The AIM2 inflammasome is critical for innate immunity to Francisella tularensis. Nat. Immunol. 11, 385–393 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Rathinam, V. A. et al. The AIM2 inflammasome is essential for host defense against cytosolic bacteria and DNA viruses. Nat. Immunol. 11, 395–402 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. McAuley, J. L. et al. Activation of the NLRP3 inflammasome by IAV virulence protein PB1-F2 contributes to severe pathophysiology and disease. PLoS Pathog. 9, e1003392 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Ichinohe, T., Pang, I. K. & Iwasaki, A. Influenza virus activates inflammasomes via its intracellular M2 ion channel. Nat. Immunol. 11, 404–410 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Lara-Tejero, M. et al. Role of the caspase-1 inflammasome in Salmonella typhimurium pathogenesis. J. Exp. Med. 203, 1407–1412 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Broz, P. et al. Redundant roles for inflammasome receptors NLRP3 and NLRC4 in host defense against Salmonella. J. Exp. Med. 207, 1745–1755 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Maltez, V. I. & Miao, E. A. Reassessing the evolutionary importance of inflammasomes. J. Immunol. 196, 956–962 (2016).

    CAS  PubMed  Google Scholar 

  64. Tsuchiya, K. et al. Caspase-1 initiates apoptosis in the absence of gasdermin D. Nat. Commun. 10, 2091 (2019). This report demonstrates that caspase 1 can induce cell killing in the absence of gasdermin D.

    PubMed  PubMed Central  Google Scholar 

  65. Miao, E. A. et al. Caspase-1-induced pyroptosis is an innate immune effector mechanism against intracellular bacteria. Nat. Immunol. 11, 1136–1142 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Brown, A. S. et al. Cooperation between monocyte-derived cells and lymphoid cells in the acute response to a bacterial lung pathogen. PLoS Pathog. 12, e1005691 (2016).

    PubMed  PubMed Central  Google Scholar 

  67. Mascarenhas, D. P. A. et al. Inhibition of caspase-1 or gasdermin-D enable caspase-8 activation in the Naip5/NLRC4/ASC inflammasome. PLoS Pathog. 13, e1006502 (2017).

    PubMed  PubMed Central  Google Scholar 

  68. Stewart, M. K. & Cookson, B. T. Evasion and interference: intracellular pathogens modulate caspase-dependent inflammatory responses. Nat. Rev. Microbiol. 14, 346–359 (2016).

    CAS  PubMed  Google Scholar 

  69. Maltez, V. I. et al. Inflammasomes coordinate pyroptosis and natural killer cell cytotoxicity to clear infection by a ubiquitous environmental bacterium. Immunity 43, 987–997 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Wong Fok Lung, T., Pearson, J. S., Schuelein, R. & Hartland, E. L. The cell death response to enteropathogenic Escherichia coli infection. Cell. Microbiol. 16, 1736–1745 (2014).

    PubMed  Google Scholar 

  71. Pearson, J. S. et al. A type III effector antagonizes death receptor signalling during bacterial gut infection. Nature 501, 247–251 (2013). This study identifies a process employed by bacteria to inhibit death receptor-induced apoptosis.

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Philip, N. H. et al. Caspase-8 mediates caspase-1 processing and innate immune defense in response to bacterial blockade of NF-κB and MAPK signaling. Proc. Natl Acad. Sci. USA 111, 7385–7390 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Weng, D. et al. Caspase-8 and RIP kinases regulate bacteria-induced innate immune responses and cell death. Proc. Natl Acad. Sci. USA 111, 7391–7396 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Wang, X. et al. Direct activation of RIP3/MLKL-dependent necrosis by herpes simplex virus 1 (HSV-1) protein ICP6 triggers host antiviral defense. Proc. Natl Acad. Sci. USA 111, 15438–15443 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Cho, Y. S. et al. Phosphorylation-driven assembly of the RIP1–RIP3 complex regulates programmed necrosis and virus-induced inflammation. Cell 137, 1112–1123 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Zhang, T. et al. Influenza virus Z-RNAs induce ZBP1-mediated necroptosis. Cell 180, 1115–1129.e1113 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Dondelinger, Y. et al. RIPK3 contributes to TNFR1-mediated RIPK1 kinase-dependent apoptosis in conditions of cIAP1/2 depletion or TAK1 kinase inhibition. Cell Death Differ. 20, 1381–1392 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Alvarez-Diaz, S. et al. The pseudokinase MLKL and the kinase RIPK3 have distinct roles in autoimmune disease caused by loss of death-receptor-induced apoptosis. Immunity 45, 513–526 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Nogusa, S. et al. RIPK3 activates parallel pathways of MLKL-driven necroptosis and FADD-mediated apoptosis to protect against influenza a virus. Cell Host Microbe 20, 13–24 (2016). RIPK3-mediated initiation of two distinct cell death pathways can be used for protection against viral infection.

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Rodrigue-Gervais, I. G. et al. Cellular inhibitor of apoptosis protein cIAP2 protects against pulmonary tissue necrosis during influenza virus infection to promote host survival. Cell Host Microbe 15, 23–35 (2014).

    CAS  PubMed  Google Scholar 

  81. Kuriakose, T. et al. ZBP1/DAI is an innate sensor of influenza virus triggering the NLRP3 inflammasome and programmed cell death pathways. Sci. Immunol. 1, aag2045 (2016).

    PubMed  PubMed Central  Google Scholar 

  82. Shan, B., Pan, H., Najafov, A. & Yuan, J. Necroptosis in development and diseases. Genes Dev. 32, 327–340 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Tsujimoto, Y., Gorham, J., Cossman, J., Jaffe, E. & Croce, C. M. The t(14;18) chromosome translocations involved in B-cell neoplasms result from mistakes in VDJ joining. Science 229, 1390–1393 (1985).

    CAS  PubMed  Google Scholar 

  84. Vaux, D. L., Cory, S. & Adams, J. M. Bcl-2 gene promotes haemopoietic cell survival and cooperates with c-myc to immortalize pre-B cells. Nature 335, 440–442 (1988). This report identifies the function of BCL-2.

    CAS  PubMed  Google Scholar 

  85. Beroukhim, R. et al. The landscape of somatic copy-number alteration across human cancers. Nature 463, 899–905 (2010). This study demonstrates somatic copy number amplifications of the genomic regions that encode MCL-1 or BCL-XL in a substantial fraction of human cancers.

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Strasser, A., Harris, A. W., Bath, M. L. & Cory, S. Novel primitive lymphoid tumours induced in transgenic mice by cooperation between myc and bcl-2. Nature 348, 331–333 (1990). Strasser et al. provide the first demonstration that blocking apoptosis promotes the development of cancer.

    CAS  PubMed  Google Scholar 

  87. Egle, A., Harris, A. W., Bouillet, P. & Cory, S. Bim is a suppressor of Myc-induced mouse B cell leukemia. Proc. Natl Acad. Sci. USA 101, 6164–6169 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Michalak, E. M. et al. Puma and to a lesser extent Noxa are suppressors of Myc-induced lymphomagenesis. Cell Death Differ. 16, 684–696 (2009).

    CAS  PubMed  Google Scholar 

  89. Strasser, A., Harris, A. W. & Cory, S. Em-bcl-2 transgene facilitates spontaneous transformation of early pre-B and immunoglobulin-secreting cells but not T cells. Oncogene 8, 1–9 (1993).

    CAS  PubMed  Google Scholar 

  90. Strasser, A., Harris, A. W., Jacks, T. & Cory, S. DNA damage can induce apoptosis in proliferating lymphoid cells via p53-independent mechanisms inhibitable by Bcl-2. Cell 79, 329–339 (1994).

    CAS  PubMed  Google Scholar 

  91. Newton, K. & Strasser, A. Ionizing radiation and chemotherapeutic drugs induce apoptosis in lymphocytes in the absence of fas or FADD/MORT1 signaling: implications for cancer therapy. J. Exp. Med. 191, 195–200 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Galluzzi, L., Buque, A., Kepp, O., Zitvogel, L. & Kroemer, G. Immunogenic cell death in cancer and infectious disease. Nat. Rev. Immunol. 17, 97–111 (2017).

    CAS  PubMed  Google Scholar 

  93. Villunger, A. et al. p53- and drug-induced apoptotic responses mediated by BH3-only proteins puma and noxa. Science 302, 1036–1038 (2003).

    CAS  PubMed  Google Scholar 

  94. Jeffers, J. R. et al. Puma is an essential mediator of p53-dependent and -independent apoptotic pathways. Cancer Cell 4, 321–328 (2003). Together with Villunger et al. (2003), this study demonstrates that PUMA directly transcriptionally activated by p53 is critical for the killing of cells by DNA damage-inducing anticancer agents.

    CAS  PubMed  Google Scholar 

  95. 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). Bouillet et al. provide the first demonstration that BH3-only proteins are essential for the initiation of apoptosis.

    CAS  PubMed  Google Scholar 

  96. Cragg, M. S., Harris, C., Strasser, A. & Scott, C. L. Unleashing the power of inhibitors of oncogenic kinases through BH3 mimetics. Nat. Rev. Cancer 9, 321–326 (2009).

    CAS  PubMed  Google Scholar 

  97. Oltersdorf, T. et al. An inhibitor of Bcl-2 family proteins induces regression of solid tumours. Nature 435, 677–681 (2005). This study describes the first BH3-mimetic drug for cancer therapy.

    CAS  PubMed  Google Scholar 

  98. Merino, D. et al. BH3-mimetic drugs: blazing the trail for new cancer medicines. Cancer Cell 34, 879–891 (2018).

    CAS  PubMed  Google Scholar 

  99. Montero, J. & Letai, A. Why do BCL-2 inhibitors work and where should we use them in the clinic? Cell Death Differ. 25, 56–64 (2018).

    CAS  PubMed  Google Scholar 

  100. Roberts, A. W. et al. Targeting BCL2 with venetoclax in relapsed chronic lymphocytic leukemia. N. Engl. J. Med. 374, 311–322 (2016). Roberts et al. present clinical trial data from using the BCL-2 inhibitor venetoclax in CLL, which led to FDA approval of this drug.

    CAS  PubMed  Google Scholar 

  101. Kelly, G. L. et al. Targeting of MCL-1 kills MYC-driven mouse and human lymphomas even when they bear mutations in p53. Genes Dev. 28, 58–70 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Kotschy, A. et al. The MCL1 inhibitor S63845 is tolerable and effective in diverse cancer models. Nature 538, 477–482 (2016).

    PubMed  Google Scholar 

  103. Brennan, M. S. et al. Humanized Mcl-1 mice enable accurate preclinical evaluation of MCL-1 inhibitors destined for clinical use. Blood 132, 1573–1583 (2018).

    CAS  PubMed  Google Scholar 

  104. Seifert, L. et al. The necrosome promotes pancreatic oncogenesis via CXCL1 and Mincle-induced immune suppression. Nature 532, 245–249 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Wang, W. et al. RIP1 kinase drives macrophage-mediated adaptive immune tolerance in pancreatic cancer. Cancer Cell 34, 757–774 e757 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  106. Patel, S. et al. RIP1 inhibition blocks inflammatory diseases but not tumor growth or metastases. Cell Death Differ. 27, 161–175 (2020).

    CAS  PubMed  Google Scholar 

  107. Baker, K. J., Houston, A. & Brint, E. IL-1 family members in cancer; two sides to every story. Front. Immunol. 10, 1197 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  108. Grivennikov, S. I., Greten, F. R. & Karin, M. Immunity, inflammation, cancer. Cell 140, 883–899 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Wang, Q. et al. A bioorthogonal system reveals antitumour immune function of pyroptosis. Nature 579, 421–426 (2020). This study demonstrates the potential utility of activators of pyroptosis in cancer therapy.

    CAS  PubMed  Google Scholar 

  110. Zhang, Z. et al. Gasdermin E suppresses tumour growth by activating anti-tumour immunity. Nature 579, 415–420 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  111. Wang, Y. et al. Chemotherapy drugs induce pyroptosis through caspase-3 cleavage of a gasdermin. Nature 547, 99–103 (2017). This article demonstrates that caspase-3-mediated cleavage of a gasdermin causes pyroptotic death in cells treated with chemotherapeutic drugs.

    CAS  PubMed  Google Scholar 

  112. Liu, Y. et al. Gasdermin E-mediated target cell pyroptosis by CAR T cells triggers cytokine release syndrome. Sci. Immunol. 5, eaax7969 (2020).

    CAS  PubMed  Google Scholar 

  113. Yuan, J., Amin, P. & Ofengeim, D. Necroptosis and RIPK1-mediated neuroinflammation in CNS diseases. Nat. Rev. Neurosci. 20, 19–33 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Caccamo, A. et al. Necroptosis activation in Alzheimer’s disease. Nat. Neurosci. 20, 1236–1246 (2017).

    CAS  PubMed  Google Scholar 

  115. Ito, Y. et al. RIPK1 mediates axonal degeneration by promoting inflammation and necroptosis in ALS. Science 353, 603–608 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  116. Heneka, M. T., McManus, R. M. & Latz, E. Inflammasome signalling in brain function and neurodegenerative disease. Nat. Rev. Neurosci. 19, 610–621 (2018).

    CAS  PubMed  Google Scholar 

  117. Ising, C. et al. NLRP3 inflammasome activation drives tau pathology. Nature 575, 669–673 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  118. Rohn, T. T. The role of caspases in Alzheimer’s disease; potential novel therapeutic opportunities. Apoptosis 15, 1403–1409 (2010).

    CAS  PubMed  Google Scholar 

  119. Paradis, E., Douillard, H., Koutroumanis, M., Goodyer, C. & LeBlanc, A. Amyloid β peptide of Alzheimer’s disease downregulates Bcl-2 and upregulates bax expression in human neurons. J. Neurosci. 16, 7533–7539 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  120. Kitamura, Y. et al. Alteration of proteins regulating apoptosis, Bcl-2, Bcl-x, Bax, Bak, Bad, ICH-1 and CPP32, in Alzheimer’s disease. Brain Res. 780, 260–269 (1998).

    CAS  PubMed  Google Scholar 

  121. Graham, S. H., Chen, J. & Clark, R. S. Bcl-2 family gene products in cerebral ischemia and traumatic brain injury. J. Neurotrauma 17, 831–841 (2000).

    CAS  PubMed  Google Scholar 

  122. Del, Re,D. P., Amgalan, D., Linkermann, A., Liu, Q. & Kitsis, R. N. Fundamental mechanisms of regulated cell death and implications for heart disease. Physiol. Rev. 99, 1765–1817 (2019).

    Google Scholar 

  123. Newton, K. et al. RIPK3 deficiency or catalytically inactive RIPK1 provides greater benefit than MLKL deficiency in mouse models of inflammation and tissue injury. Cell Death Differ. 23, 1565–1576 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  124. Fang, X. et al. Ferroptosis as a target for protection against cardiomyopathy. Proc. Natl Acad. Sci. USA 116, 2672–2680 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  125. Voet, S., Srinivasan, S., Lamkanfi, M. & van Loo, G. Inflammasomes in neuroinflammatory and neurodegenerative diseases. EMBO Mol. Med. 11, e10248 (2019).

    PubMed  PubMed Central  Google Scholar 

  126. Varfolomeev, E. E. et al. Targeted disruption of the mouse caspase 8 gene ablates cell death induction by the TNF receptors, Fas/Apo1, and DR3 and is lethal prenatally. Immunity 9, 267–276 (1998).

    CAS  PubMed  Google Scholar 

  127. Newton, K., Harris, A. W., Bath, M. L., Smith, K. G. C. & Strasser, A. A dominant interfering mutant of FADD/Mort1 enhances deletion of autoreactive thymocytes and inhibits proliferation of mature T lymphocytes. EMBO J. 17, 706–718 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  128. Kaiser, W. J. et al. RIP3 mediates the embryonic lethality of caspase-8-deficient mice. Nature 471, 368–372 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  129. Oberst, A. et al. Catalytic activity of the caspase-8-FLIP(L) complex inhibits RIPK3-dependent necrosis. Nature 471, 363–367 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  130. Guasparri, I., Keller, S. A. & Cesarman, E. KSHV vFLIP is essential for the survival of infected lymphoma cells. J. Exp. Med. 199, 993–1003 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  131. Turner, S., Kenshole, B. & Ruby, J. Viral modulation of the host response via crmA/SPI-2 expression. Immunol. Cell Biol. 77, 236–241 (1999).

    CAS  PubMed  Google Scholar 

  132. He, S. & Han, J. Manipulation of host cell death pathways by herpes simplex virus. Curr. Top. Microbiol. Immunol. (2020).

  133. Pauleau, A. L. et al. Structure-function analysis of the interaction between Bax and the cytomegalovirus-encoded protein vMIA. Oncogene 26, 7067–7080 (2007).

    CAS  PubMed  Google Scholar 

  134. Wang, T. et al. Necroptosis is dispensable for motor neuron degeneration in a mouse model of ALS. Cell Death Differ. 27, 1728–1739 (2020).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  136. Fritsch, M. et al. Caspase-8 is the molecular switch for apoptosis, necroptosis and pyroptosis. Nature 575, 683–687 (2019).

    CAS  PubMed  Google Scholar 

  137. Newton, K. et al. Activity of caspase-8 determines plasticity between cell death pathways. Nature 575, 679–682 (2019). Together with Fritsch et al. (2019), this article demonstrates the ability of caspase-8 to coordinate the activation of several distinct cell death pathways.

    CAS  PubMed  Google Scholar 

  138. Schwarzer, R., Jiao, H., Wachsmuth, L., Tresch, A. & Pasparakis, M. FADD and caspase-8 regulate gut homeostasis and inflammation by controlling MLKL- and GSDMD-mediated death of intestinal epithelial cells. Immunity 52, 978–993.e6 (2020).

    CAS  PubMed  Google Scholar 

  139. Tummers, B. et al. Caspase-8-dependent inflammatory responses are controlled by its adaptor, FADD, and necroptosis. Immunity 52, 994–1006.e8 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  140. Chen, K. W. et al. Extrinsic and intrinsic apoptosis activate pannexin-1 to drive NLRP3 inflammasome assembly. EMBO J. 38, e101638 (2019).

    PubMed  PubMed Central  Google Scholar 

  141. Vince, J. E. et al. The mitochondrial apoptotic effectors BAX/BAK activate caspase-3 and -7 to trigger NLRP3 inflammasome and caspase-8 driven IL-1β activation. Cell Rep. 25, 2339–2353 (2018).

    CAS  PubMed  Google Scholar 

  142. Lampson, B. L. & Davids, M. S. The development and current use of BCL-2 inhibitors for the treatment of chronic lymphocytic leukemia. Curr. Hematol. Malig. Rep. 12, 11–19 (2017).

    PubMed  PubMed Central  Google Scholar 

  143. Marsden, V. et al. Apoptosis initiated by Bcl-2-regulated caspase activation independently of the cytochrome c/Apaf-1/caspase-9 apoptosome. Nature 419, 634–637 (2002).

    CAS  PubMed  Google Scholar 

  144. Bock, F. J. & Tait, S. W. G. Mitochondria as multifaceted regulators of cell death. Nat. Rev. Mol. Cell Biol. 21, 85–100 (2020).

    CAS  PubMed  Google Scholar 

  145. Vallabhapurapu, S. & Karin, M. Regulation and function of NF-κB transcription factors in the immune system. Annu. Rev. Immunol. 27, 693–733 (2009).

    CAS  PubMed  Google Scholar 

  146. Iwai, K. & Tokunaga, F. Linear polyubiquitination: a new regulator of NF-κB activation. EMBO Rep. 10, 706–713 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  147. Shim, J. H. et al. TAK1, but not TAB1 or TAB2, plays an essential role in multiple signaling pathways in vivo. Genes Dev. 19, 2668–2681 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  148. Chen, G. & Goeddel, D. V. TNF-R1 signaling: a beautiful pathway. Science 296, 1634–1635 (2002).

    CAS  PubMed  Google Scholar 

  149. Grossmann, M. et al. The anti-apoptotic activities of rel and RelA required during B-cell maturation involve the regulation of Bcl-2 expression. EMBO J. 19, 6351–6360 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  150. Baldwin, A. S. Regulation of cell death and autophagy by IKK and NF-κB: critical mechanisms in immune function and cancer. Immunol. Rev. 246, 327–345 (2012).

    PubMed  Google Scholar 

  151. Meinzer, U. et al. Yersinia pseudotuberculosis effector YopJ subverts the Nod2/RICK/TAK1 pathway and activates caspase-1 to induce intestinal barrier dysfunction. Cell Host Microbe 11, 337–351 (2012).

    CAS  PubMed  Google Scholar 

  152. Mukherjee, S. et al. Yersinia YopJ acetylates and inhibits kinase activation by blocking phosphorylation. Science 312, 1211–1214 (2006).

    CAS  PubMed  Google Scholar 

  153. Peltzer, N. & Walczak, H. Cell Death and inflammation — a vital but dangerous Liaison. Trends Immunol. 40, 387–402 (2019).

    CAS  PubMed  Google Scholar 

  154. Lei, X. et al. Enterovirus 71 3C inhibits cytokine expression through cleavage of the TAK1/TAB1/TAB2/TAB3 complex. J. Virol. 88, 9830–9841 (2014).

    PubMed  PubMed Central  Google Scholar 

  155. Zhou, X. et al. A Vibrio parahaemolyticus T3SS effector mediates pathogenesis by independently enabling intestinal colonization and inhibiting TAK1 activation. Cell Rep. 3, 1690–1702 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  156. He, C. et al. Bacterial nucleotidyl cyclase inhibits the host innate immune response by suppressing TAK1 activation. Infect. Immun. 85, e00239-17 (2017).

    PubMed  PubMed Central  Google Scholar 

  157. Nilsson, J. A. & Cleveland, J. L. Myc pathways provoking cell suicide and cancer. Oncogene 22, 9007–9021 (2003).

    CAS  PubMed  Google Scholar 

  158. Fanidi, A., Harrington, E. A. & Evan, G. I. Cooperative interaction between c-myc and bcl-2 proto-oncogenes. Nature 359, 554–556 (1992).

    CAS  PubMed  Google Scholar 

  159. Bissonnette, R. P., Echeverri, F., Mahboubi, A. & Green, D. R. Apoptotic cell death induced by c-myc is inhibited by bcl-2. Nature 359, 552–554 (1992).

    CAS  PubMed  Google Scholar 

  160. Gong, J. N. et al. Hierarchy for targeting prosurvival BCL2 family proteins in multiple myeloma: pivotal role of MCL1. Blood 128, 1834–1844 (2016).

    CAS  PubMed  Google Scholar 

  161. Nassour, J. et al. Autophagic cell death restricts chromosomal instability during replicative crisis. Nature 565, 659–663 (2019). Nassour et al. demonstrate the role of autophagic cell death in the removal of cells with chromosomal instability.

    CAS  PubMed  PubMed Central  Google Scholar 

  162. Poillet-Perez, L. & White, E. Role of tumor and host autophagy in cancer metabolism. Genes Dev. 33, 610–619 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  163. Yamamoto, K. et al. Autophagy promotes immune evasion of pancreatic cancer by degrading MHC-I. Nature 581, 100–105 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  164. Ofengeim, D. et al. RIPK1 mediates a disease-associated microglial response in Alzheimer’s disease. Proc. Natl Acad. Sci. USA 114, E8788–E8797 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  165. Taabazuing, C. Y., Okondo, M. C. & Bachovchin, D. A. Pyroptosis and apoptosis pathways engage in bidirectional crosstalk in monocytes and macrophages. Cell Chem. Biol. 24, 507–514.e504 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  166. Man, S. M. et al. Salmonella infection induces recruitment of caspase-8 to the inflammasome to modulate IL-1β production. J. Immunol. 191, 5239–5246 (2013).

    CAS  PubMed  Google Scholar 

  167. Van Opdenbosch, N. et al. Caspase-1 engagement and TLR-induced c-FLIP expression suppress ASC/caspase-8-dependent apoptosis by inflammasome sensors NLRP1β and NLRC4. Cell Rep. 21, 3427–3444 (2017).

    PubMed  PubMed Central  Google Scholar 

  168. Chung, H. et al. NLRP3 regulates a non-canonical platform for caspase-8 activation during epithelial cell apoptosis. Cell Death Differ. 23, 1331–1346 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  169. Doerflinger, M. et al. Flexible usage and interconnectivity of diverse cell death pathways protects against intracellular infection. Immunity https://doi.org/10.1016/j.immuni.2020.07.004 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  170. Heilig, R. et al. Caspase-1 cleaves Bid to release mitochondrial SMAC and drive secondary necrosis in the absence of GSDMD. Life Sci. Alliance 3, e202000735 (2020).

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  172. Muendlein, H. I. et al. cFLIPL protects macrophages from LPS-induced pyroptosis via inhibition of complex II formation. Science 367, 1379–1384 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  173. Kesavardhana, S., Subbarao Malireddi, R. K. & Kanneganti, T. D. Caspases in cell death, inflammation, and gasdermin-induced pyroptosis. Annu. Rev. Immunol. 38, 567–595 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  174. Rauch, I. et al. NAIP–NLRC4 inflammasomes coordinate intestinal epithelial cell expulsion with eicosanoid and IL-18 release via activation of caspase-1 and -8. Immunity 46, 649–659 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  175. Hausmann, A. et al. Intestinal epithelial NAIP/NLRC4 restricts systemic dissemination of the adapted pathogen Salmonella Typhimurium due to site-specific bacterial PAMP expression. Mucosal Immunol. 13, 530–544 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  176. Strasser, A. & Vaux, D. L. Cell death in the origin and treatment of cancer. Mol. Cell 78, 1045–1054 (2020).

    CAS  PubMed  Google Scholar 

  177. Ascierto, P. A. et al. Future perspectives in melanoma research ‘Melanoma Bridge’, Napoli, November 30th–3rd December 2016. J. Transl Med. 15, 236 (2017).

    PubMed  PubMed Central  Google Scholar 

  178. Wei, S. C., Duffy, C. R. & Allison, J. P. Fundamental mechanisms of immune checkpoint blockade therapy. Cancer Discov. 8, 1069–1086 (2018).

    PubMed  Google Scholar 

  179. Miller, J. F. & Sadelain, M. The journey from discoveries in fundamental immunology to cancer immunotherapy. Cancer Cell 27, 439–449 (2015).

    CAS  PubMed  Google Scholar 

  180. Speir, M. et al. Eliminating Legionella by inhibiting BCL-XL to induce macrophage apoptosis. Nat. Microbiol. 1, 15034 (2016).

    CAS  PubMed  Google Scholar 

  181. Kostic, V., Jackson-Lewis, V., de Bilbao, F., Dubois-Dauphin, M. & Przedborski, S. Bcl-2: prolonging life in a transgenic mouse model of familial amyotrophic lateral sclerosis. Science 277, 559–562 (1997).

    CAS  PubMed  Google Scholar 

  182. Kroemer, G. & Levine, B. Autophagic cell death: the story of a misnomer. Nat. Rev. Mol. Cell Biol. 9, 1004–1010 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  183. Holze, C. et al. Oxeiptosis, a ROS-induced caspase-independent apoptosis-like cell-death pathway. Nat. Immunol. 19, 130–140 (2018).

    CAS  PubMed  Google Scholar 

  184. Dixon, S. J. et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell 149, 1060–1072 (2012). This study is the first demonstration of the mechanism of programmed cell death by ferroptosis.

    CAS  PubMed  PubMed Central  Google Scholar 

  185. Brinkmann, V. et al. Neutrophil extracellular traps kill bacteria. Science 303, 1532–1535 (2004).

    CAS  PubMed  Google Scholar 

  186. Martinez-Lostao, L., Anel, A. & Pardo, J. How do cytotoxic lymphocytes kill cancer cells? Clin. Cancer Res. 21, 5047–5056 (2015).

    CAS  PubMed  Google Scholar 

  187. Lowin, B., Peitsch, M. C. & Tschopp, J. Perforin and granzymes: crucial effector molecules in cytolytic T lymphocyte and natural killer cell-mediated cytotoxicity. Curr. Top. Microbiol. Immunol. 198, 1–24 (1995).

    CAS  PubMed  Google Scholar 

  188. Waterhouse, N. J. et al. A central role for Bid in granzyme B-induced apoptosis. J. Biol. Chem. 280, 4476–4482 (2005).

    CAS  PubMed  Google Scholar 

  189. Lopez, J. A. et al. Perforin forms transient pores on the target cell plasma membrane to facilitate rapid access of granzymes during killer cell attack. Blood 121, 2659–2668 (2013).

    CAS  PubMed  Google Scholar 

  190. Li, X., McKinstry, K. K., Swain, S. L. & Dalton, D. K. IFN-γ acts directly on activated CD4+ T cells during mycobacterial infection to promote apoptosis by inducing components of the intracellular apoptosis machinery and by inducing extracellular proapoptotic signals. J. Immunol. 179, 939–949 (2007).

    CAS  PubMed  Google Scholar 

  191. Barthson, J. et al. Cytokines tumor necrosis factor-α and interferon-γ induce pancreatic β-cell apoptosis through STAT1-mediated Bim protein activation. J. Biol. Chem. 286, 39632–39643 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We are grateful to the present and past members of our laboratories, our collaborators and the mentors that we have had the pleasure of working with. Our work is supported by grants and fellowships from the Australian National Health and Medical Research Council (Project Grants 1186575 and 1145728 to M.J.H., 1143105 to M.J.H. and A.S., 1159658 to M.J.H. and S.B., Program Grant 1016701 to A.S., and Fellowships 1020363, to A.S., and 1156095, to M.J.H.), by the Leukemia and Lymphoma Society of America (grant LLS SCOR 7001-13 to A.S. and M.J.H.), by the Cancer Council of Victoria (project grants 1147328, to M.J.H., and 1052309, to A.S.) and by a Venture Grant to M.J.H. and A.S.

Author information

Authors and Affiliations

Authors

Contributions

The authors contributed equally to all aspects of the article.

Corresponding author

Correspondence to Andreas Strasser.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information

Nature Reviews Molecular Cell Biology thanks J. Yuan, A. Oberst and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note

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

Glossary

BCL-2 (B cell leukaemia/lymphoma-2) family

A family of proteins — named after its original member, BCL-2 — that regulate the intrinsic apoptotic pathway.

Complement system

Evolutionarily ancient system of protein cascades capable of lysing bacteria by perforating their outer membrane, opsonizing the pathogens and activating the cells of the immune system.

Damage-associated molecular patterns

(DAMPs, also known as alarmins). Intracellular molecules, such as HMGB1 or S100A8, whose release from cells undergoing lytic cell death triggers distinct receptors in innate immune cells and causes inflammation.

Pathogen-associated molecular patterns

(PAMPs). Evolutionarily conserved molecular components of pathogens, such as LPS expressed by Gram-negative bacteria, that cause inflammatory responses by innate immune cells.

Pattern recognition receptors

Membrane-associated or cytosolic receptors capable of recognizing and responding to PAMPs through the induction of pro-inflammatory responses.

TAM receptors

Family of receptor tyrosine kinases (TYRO3, AXL, MERTK) that promote apoptotic cell clearance by binding to phosphatidylserine exposed on apoptotic cells using GAS6 and protein S as bridging ligands.

BH3-only proteins

Pro-apoptotic members of the BCL-2 protein family that share only one of the four BCL-2 homology (BH) domains, namely the BH3 domain, with the remainder of the family. BH3-only proteins are induced transcriptionally and/or activated post-translationally in response to developmental cues or cytotoxic stimuli that initiate the intrinsic apoptotic cell death pathway.

Mitochondrial outer membrane permeabilization

(MOMP). Perforation of the outer mitochondrial membrane, causing leakage of content from the mitochondrial intermembrane space, including the apoptosis inducers cytochrome c and SMAC. MOMP can result in the translocation of mitochondrial DNA into the cytosol, leading to the production of type I interferons and thereby driving inflammatory responses.

Inhibitor of apoptosis proteins

(IAPs). Family of proteins with structural homology (that is, baculovirus IAP repeats). Some of the IAP proteins have an E3 ubiquitin ligase function, allowing them to ubiquitylate their target proteins. XIAP inhibits apoptosis by binding to and promoting the degradation of caspases 3 and 7, whereas cIAP1 and cIAP2 promote pro-survival signalling from TNFR1 by enhancing NF-κB activation.

Death receptors

Subsets of the TNFR superfamily that contain an intracellular death domain, which upon ligation can induce killing of the cells on which they are expressed through FADD adaptor protein-mediated activation of caspase 8.

cFLIP

(cellular CASP8 and FADD-like apoptosis regulator). Protein with structural similarity to caspase 8 but that lacks enzymatic activity. There are two forms of FLIP: FLIP short and FLIP long. FLIP short inhibits apoptosis by preventing the activation of caspase 8; FLIP long can form heterodimers with caspase 8, and this heterodimer inhibits necroptosis by cleaving RIPK1. High levels of FLIP long can also inhibit caspase 8 activation and apoptosis.

Inflammasomes

Multimeric protein complexes, activated by various events, including ion flux, reactive oxygen species and mitochondrial dysfunction. They comprise sensors, such as NLR molecules, and their formation often depends on the adaptor protein ASC and pro-caspase 1, which together cause the autocatalytic activation of caspase 1. The consequent proteolytic processing of pro-IL-1β and pro-IL-18 into their bioactive forms results in inflammation and proteolytic activation of gasdermin D to drive pyroptotic cell death.

NLR (nucleotide-binding domain and leucine-rich repeat containing) family

Evolutionarily conserved diverse family of proteins, further classified into NLRA, NLRB, NLRP and NLRC, in accordance with their N-terminal domains and the presence or absence of CARD domains. Certain (but probably not all) NLRs function in innate immune sensing of pathogens and infection-associated cellular changes. They contribute to the protection of the infected host by instructing an antimicrobial defence, including inflammatory responses.

Type III secretion (T3SS) apparatus

Complex molecular machines used by bacteria to inject effector proteins into eukaryotic host cells.

Flagellin

Subunit protein of the flagellum that endows bacteria with motility.

Gasdermin family

Conserved family of proteins in vertebrates, named after the restriction of gasdermin A to gut and skin epithelial cells, although it is now clear that these proteins are much more widely expressed. At least some of the gasdermins can form pores in membranes after proteolytic cleavage (for example, through processing of gasdermin D by caspase 1 or 11).

ESCRT

(endosomal sorting complexes required for transport). Multiprotein machinery that enables membrane bending/budding away from the cytoplasm.

Lipopolysaccharide

(LPS). Large molecules, comprising a lipid and a complex polysaccharide, found in the outer membrane of Gram-negative bacteria.

Gram-negative bacteria

Diverse group of bacteria defined by their inability to retain crystal violet (or Gram) stains, because of the architecture of their cell envelope being composed of an inner cytoplasmic and outer bacterial cell membrane separated by a thin peptidoglycan cell wall. Typical examples include Escherichia coli, Salmonella enterica, Pseudomonas aeruginosa, Chlamydia trachomatis and Yersinia pestis.

Toll-like receptor

Family of transmembrane receptors that recognize PAMPs and DAMPs and, upon stimulation, can induce diverse pro-inflammatory responses.

RIG-I-like receptors

Cytosolic pattern recognition receptors that respond to double-stranded RNA.

Antigen-presenting cells

While all nucleated cells can present antigens, the group of professional antigen-presenting cells, which comprise macrophages, dendritic cells and B cells, are capable of priming naive T cells by the processing and presentation of antigen-derived peptides in the context of class I or class II MHC molecules and by the delivery of co-stimulatory signals.

PB1-F2

A protein encoded by the influenza A virus that contributes to its pathogenicity.

M2 protein

Protein encoded by the influenza A virus that is part of the viral envelope; it is capable of forming a tunnel between host cell compartments.

Z-RNAs

Left-handed form of double-stranded RNA that is bound by proteins, such as ADAR, ZBP1 or their viral homologues.

Cytotoxic lymphocytes

CD8+ T cells and natural killer cells, which can kill infected or malignant cells via diverse mechanisms, including the delivery of perforin and granzymes, the use of FAS ligand to activate the death receptor FAS, and the delivery of IFNγ to target cells.

BH3-mimetic drugs

Small-molecule inhibitors of pro-survival BCL-2 proteins. They mimic the action of the pro-apoptotic BH3-only proteins, which as the natural cellular inhibitors of pro-survival BCL-2 proteins are critical for the initiation of the intrinsic apoptosis signalling pathway.

Necrosome

Protein complex consisting of RIPK1, RIPK3 and FADD. This complex is formed in response to TNFR1 stimulation when both the activation of NF-κB and caspase 8 activity are blocked. This signalling complex causes the activation of the pseudokinase MLKL, the critical effector of necroptosis.

Tumour lysis syndrome

Caused by the failure to safely remove large numbers of dying tumour cells during anticancer therapy, which can cause renal failure, cardiac abnormalities, seizures and sudden death.

CAR (chimeric antigen receptor) T cells

T lymphocytes engineered to express artificial antigen receptors capable of directly recognizing proteins on cancer cells and killing these malignant cells.

Amyloid plaques

Beta-amyloid protein aggregates implicated in the destruction of nerve connections, thus causing degenerative disorders, such as Alzheimer disease.

Periodic fever syndrome

Group of rare genetic autoinflammatory diseases in which patients develop periodic fevers with a range of inflammatory pathologies, including stomatitis, aphtitis and adenitis.

Serpin

Superfamily of proteins that share a structural homology, where many perform serine protease inhibitory activity whereas others (for example, CrmA from cowpox virus) perform cysteine protease inhibitory activity.

Canonical and non-canonical NF-κB pathways

Induced by the stimulation of a variety of surface receptors (for example, TLRs, members of the TNFR superfamily, and antigen receptors), the two distinct NF-κB signalling pathways involve different members of the NF-κB/REL protein family. The classical/canonical NF-κB pathway operates via heterodimers of NF-κB1 (its cleavage product p50) with RELA or c-REL, whereas the non-canonical pathway is mediated mainly by heterodimers of NF-κB2 (its cleavage product p52) with RELB.

cGAS

Intracellular DNA sensor that induces an interferon response.

Mast cells

Tissue-resident cells involved in immune defence against parasitic infections and allergic responses.

Ripoptosomes

Signalling platforms comprising RIPK1, RIPK3, FADD and caspase 8 that can induce either apoptosis or necroptosis, depending on the state of the cell.

Immune checkpoint signalling

Signalling pathways that attenuate the activity of immune cells, mainly T lymphocytes, thereby regulating immune responses and preventing the destruction of self-tissues (contributing to self-tolerance). The inhibition of immune checkpoint regulators, such as PD1 or CTLA4, can enhance CD8+ cytotoxic T cell-mediated killing of cancer cells.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Bedoui, S., Herold, M.J. & Strasser, A. Emerging connectivity of programmed cell death pathways and its physiological implications. Nat Rev Mol Cell Biol 21, 678–695 (2020). https://doi.org/10.1038/s41580-020-0270-8

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41580-020-0270-8

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