key: cord-0991382-y63f0x82 authors: Gullett, Jessica M.; Tweedell, Rebecca E.; Kanneganti, Thirumala-Devi title: It’s All in the PAN: Crosstalk, Plasticity, Redundancies, Switches, and Interconnectedness Encompassed by PANoptosis Underlying the Totality of Cell Death-Associated Biological Effects date: 2022-04-29 journal: Cells DOI: 10.3390/cells11091495 sha: 899134004ae3313113802de9791c58f4eea03412 doc_id: 991382 cord_uid: y63f0x82 The innate immune system provides the first line of defense against cellular perturbations. Innate immune activation elicits inflammatory programmed cell death in response to microbial infections or alterations in cellular homeostasis. Among the most well-characterized programmed cell death pathways are pyroptosis, apoptosis, and necroptosis. While these pathways have historically been defined as segregated and independent processes, mounting evidence shows significant crosstalk among them. These molecular interactions have been described as ‘crosstalk’, ‘plasticity’, ‘redundancies’, ‘molecular switches’, and more. Here, we discuss the key components of cell death pathways and note several examples of crosstalk. We then explain how the diverse descriptions of crosstalk throughout the literature can be interpreted through the lens of an integrated inflammatory cell death concept, PANoptosis. The totality of biological effects in PANoptosis cannot be individually accounted for by pyroptosis, apoptosis, or necroptosis alone. We also discuss PANoptosomes, which are multifaceted macromolecular complexes that regulate PANoptosis. We consider the evidence for PANoptosis, which has been mechanistically characterized during influenza A virus, herpes simplex virus 1, Francisella novicida, and Yersinia infections, as well as in response to altered cellular homeostasis, in inflammatory diseases, and in cancers. We further discuss the role of IRF1 as an upstream regulator of PANoptosis and conclude by reexamining historical studies which lend credence to the PANoptosis concept. Cell death has been shown to play a critical role in infections, inflammatory diseases, neurodegenerative diseases, cancers, and more; therefore, having a holistic understanding of cell death is important for identifying new therapeutic strategies. The innate immune system is the first line of defense against infection and cellular insults; innate immune receptors can recognize the molecular signatures of pathogens, called pathogen-associated molecular patterns (PAMPs), as well as components released by damaged cells, called damage-associated molecular patterns (DAMPs). The innate immune system activates genetically defined programmed cell death pathways in response to microbial infections or alterations in cellular homeostasis; among the most well characterized of these programmed cell death responses are pyroptosis, apoptosis, and necroptosis. Though canonically proposed as segregated cellular processes responding to individualized PAMPs and DAMPs, mounting evidence shows significant interactions between the components of pyroptosis, apoptosis, and necroptosis. Historically, the literature on cell death and Among the most comprehensively studied cell death processes to date are pyroptosis, apoptosis, and necroptosis [21, 22] . Each occurs in response to cellular insults, but they differ in terms of their molecular machinery. Pyroptosis is a lytic form of proinflammatory cell death that was originally described as a caspase-1-mediated death [23] . Pyroptotic cell death typically involves the formation of the inflammasome, a supramolecular platform that is composed of a sensor, adaptor protein ASC, and caspase-1 [24] . The five most well-known inflammasomes, which are named after their corresponding sensor based on the genetic characterization of sensors and triggers, are the NLR family inflammasomes, NLRP1 [24] , NLRP3 [25] [26] [27] , and NAIP/NLRC4 [28] [29] [30] , as well as those formed by other sensors containing pyrin domains, such as Pyrin [31] and AIM2 [32, 33] . Sensor activation polymerizes the adaptor protein ASC into prion-like structures referred to as ASC specks [34] [35] [36] , which recruit caspase-1 to allow its autoproteolysis and activation [24, 37] . Activated caspase-1 cleaves inflammatory cytokines IL-1β and IL-18 [38] as well as the pore-forming molecule gasdermin D (GSDMD) [39] . The GSDMD-mediated pores allow the release of the inflammatory cytokines [40] [41] [42] [43] [44] along with other inflammatory molecules such as HMGB1, which serve as DAMPs and further propagate an innate immune inflammatory response. GSDMD is also activated by caspase-11 (mice) and caspase-4/5 (humans) in the process of non-canonical inflammasome activation [39] [40] [41] 44, 45] . In addition to requiring cleavage for activation, GSDMD is also regulated at the transcriptional level by IFN regulatory factor 2 (IRF2), with a compensatory role for IRF1, in murine bone marrow-derived macrophages (BMDMs) [46] ; in human cells IRF2 does not regulate GSDMD expression but does regulate caspase-4-mediated cell death, and IRF1 acts cooperatively in this process in response to IFN-γ [47] . Apoptosis is a form of programmed cell death originally described as a 'mechanism of controlled cell deletion' characterized by its distinct morphological membrane blebbing and subsequent cell shrinkage [48] . It proceeds through either an extrinsic or intrinsic pathway, though both result in the activation of the same executioner caspases. The intrinsic pathway forms an APAF1-mediated apoptosome in response to homeostatic disruptions, such as DNA damage or loss of mitochondrial stability [49] . This multiprotein complex includes APAF1, cytochrome c, and the initiator caspase caspase-9 which, upon cleavage, activates the downstream effector/executioner caspases, caspase-3 and -7 [50, 51] . Extrinsic apoptosis occurs after ligand binding to death receptors, such as Fas and TNF-α receptor (TNFR), on the cell surface; downstream of death receptor binding, FADD translocates to the receptor, which recruits caspase-8. Caspase-8 is the key extrinsic apoptotic initiator caspase which cleaves downstream caspases, caspase-3 and -7, to execute cell death [52, 53] . Caspase-8 can also induce activation of intrinsic apoptosis by activating the proapoptotic molecule Bid [54] [55] [56] , which translocates to the mitochondria to facilitate pore formation by BAX/BAK and induce mitochondrial outer membrane permeabilization (MOMP) and apoptosome formation [57, 58] . Necroptosis, another lytic form of cell death, occurs in response to caspase-8 inhibition and is RIPK3-and MLKL-dependent [22, [59] [60] [61] . The apoptotic caspase-8 typically blocks necroptosis by cleaving RIPK3, CYLD, and RIPK1 [62] [63] [64] . Necroptosis can be initiated in response to the activation of toll-like receptors (TLRs), death receptors, or through interferon (IFN) signaling [65] . A well-characterized necroptosis response is induced by TNF-α. Its binding to TNFR induces signaling that activates RIPK1 to become phosphorylated and, along with TRADD, FADD, and caspase-8, form complex II [66] . When caspase-8 is inhibited, RIPK1 interacts with RIPK3 to form a cell death-inducing necrosome. The RIPK1-RIPK3 complex promotes phosphorylation of MLKL, causing its oligomerization. The MLKL multimer then translocates to the plasma membrane, where it interacts with phospholipids and forms pores [67, 68] . In addition to MLKL phosphorylation by RIPK3, MLKL activity is also regulated by other post-translational modifications, such as ubiquitylation, which is necessary for higher-order oligomerization [69] . Within each of these cell death pathways, there are several regulators and auxiliary components. For example, additional inflammasome components have been identified that are involved in its regulation and activation, such as NEK7 [70] [71] [72] [73] and DDX3X [74] , as well as transcription factors such as IRF1 [75] , IRF2 [46, 47] , and IRF8 [76] . Additionally, NINJ1 has been identified as a critical component for plasma membrane rupture [77] . There are also variations in the signaling cascades that exist within each programmed cell death pathway, making the complexity of these cellular processes, including regulatory components, cell-and trigger-specific responses, and time-dependent responses, limitless. Additional components and layers of complexity have been extensively reviewed elsewhere [22, 78, 79] . Understanding the activation and execution of inflammatory cell death pathways has been an active area of research, particularly given the clinical relevance of cell death pathways in infections, inflammatory diseases, cancers, and beyond [2, 4, 5, [7] [8] [9] [11] [12] [13] 16, [18] [19] [20] . As a result of these studies, several examples of crosstalk and flexibility have been identified between the molecular components of programmed cell death pathways. Here, we will limit our discussion to genetically defined examples over time. At their core, apoptosis and necroptosis are intricately molecularly linked, given that TNF-induced caspase-8 activation drives apoptosis while inhibition of caspase-8 during this process drives necroptosis [80] . The rescue of caspase-8-deficient embryos by the loss of RIPK3 or MLKL has long been documented [81] [82] [83] [84] , and enzymatically active caspase-8 is critical in the regulation and balance of apoptosis and necroptosis [85, 86] . Beyond the intrinsic connection between apoptosis and necroptosis, caspase-1, an essential component of inflammasomes, cleaves apoptosis-associated caspase-7 during Salmonella infection (NLRC4 inflammasome trigger) as well as in response to LPS + ATP stimulation (NLRP3 inflammasome trigger) [17] . The pyroptotic caspase-1 also cleaves apoptotic PARP1 in response to inflammasome-activating triggers [6] , and loss of caspase-1 during Salmonella infection leads to activation of apoptotic proteins instead [87] . In addition, cells lacking pyroptotic caspase-1 and caspase-11 have reduced mitochondrial damage in response to inflammasome-activating triggers such as the NLRP3-activating LPS + ATP treatment or AIM2-activating dsDNA transfection [88] , suggesting additional crosstalk between inflammasomes and apoptotic processes. Reciprocally, the apoptotic caspase-8 serves as a regulatory component of pyroptotic inflammasomes [19] . Fluorescence microscopy has shown the colocalization of caspase-8 and ASC in both pyroptosis-deficient and pyroptosissufficient cells in response to infections [12, [89] [90] [91] . Additionally, caspase-8, along with FADD, is required to both prime and activate canonical (ligand-induced) and noncanonical (E. colior Citrobacter rodentium-induced) NLRP3 inflammasomes [19] . Caspase-8 can be recruited during NLRC4 and NLRP1b inflammasome formation [91] [92] [93] and at ASC specks involving multiple inflammasome sensors, such as NLRP3 and NLRC4 or AIM2 and Pyrin [12, 94] ; FADD can also be recruited to these ASC specks in response to FlaTox, a combination of the bacterial PAMPs Bacillus anthracis protective antigen and the N-terminus of lethal factor fused to Legionella pneumophila flagellin [93] . However, caspase-8 is not required for Salmonella-induced cell death at 2, 6, and 24 h post-infection using an MOI of 1 or 10 [91] , showcasing the variability of the roles of caspase-8 within the programmed cell death response. Crosstalk has also been identified between cell death molecules by studying the totality of biological effects in disease processes. For example, inflammatory bone disease in mice carrying the Pstpip2 cmo mutation persists despite deletion of caspase-1 or combined deletion of caspase-8/RIPK3 (deletion of caspase-8 alone is embryonically lethal [84] ); the inflammation is only rescued by the combined deletion of NLRP3 or caspase-1 with caspase-8/RIPK3 [16, 18] , highlighting the functional redundancies of pyroptotic molecules NLRP3 and caspase-1 with the apoptosis-necroptosis modulator caspase-8. In the context of infection, influenza A virus (IAV) induces activation of pyroptotic, apoptotic, and necroptotic proteins, and loss of RIPK3 protects against much of the cell death, but combined deletion of caspase-8 and RIPK3 is necessary to further reduce cell death [9] , providing additional mechanistic evidence of overlaps in the functions of molecules involved in cell death activation. Beyond caspase-8 and RIPK3, the necroptotic molecule MLKL has also been implicated in crosstalk between cell death pathways. For example, ASC oligomerization to induce NLRP3 inflammasome activation can occur in response to treatment with TLR3 ligands and zVAD, but the ASC oligomerization is blocked in MLKL-deficient cells [95] . As oligomerized MLKL forms pores in the plasma membrane, a cascade of cellular consequences begins, including the efflux of potassium ions. This necroptosis-induced ionic efflux has been shown to activate the NLRP3 inflammasome [96, 97] . Together, these data show how necroptosis and inflammasomes (pyroptosis) are interconnected. Given the recently identified role of gasdermins in cell death, it has also been found that gasdermins mediate crosstalk between cell death pathways. GSDMD was initially identified as an executioner of pyroptotic cell death in response to caspase-1, caspase-4/-5 (human) or caspase-11 (mouse) cleavage [39, 45] . Caspase-8 can also cleave GSDMD to activate pore formation and cell death during Yersinia infection [3, [98] [99] [100] . Further studies have found that GSDMD can also be processed by the apoptosis-inducing caspase-3 in such a manner that renders GSDMD inactive, suppressing pyroptosis [101] . However, inflammasome and GSDMD activation in response to Shiga toxin 2 and LPS are also associated with increased mitochondrial ROS [102] , and GSDMD can form pores in the mitochondrial membrane to release canonically proapoptotic molecules and activate caspase-3 in a BAK/BAX-independent manner [103, 104] . Other members of the gasdermin family are also increasingly implicated in cell death crosstalk. Microarray and subsequent pathway analysis of inner ear samples from day-0 postnatal mice showed that the gene set involved in apoptosis is downregulated in mice lacking Gsdme as compared with wild-type controls [105] . Furthermore, GSDME can be cleaved by caspase-3, an apoptotic cell death effector, and can induce pyroptotic death [106, 107] . In THP-1 cells lacking GSDMD, GSDME allows the release of IL-1β in response to nigericin, Val-boroPro, or Salmonella infection, though limited cell death was observed with endogenous GSDME expression levels in these cells [108] . In murine cells, NLRP3 inflammasome activation in GSDMD-deficient cells results in IL-1β and IL-18 release through caspase-8/-3 and GSDME activation [109] . GSDME serves in a feed-forward loop to promote caspase-3 activation by forming pores in the mitochondrial membrane and inducing the release of cytochrome c in response to traditional intrinsic and extrinsic apoptotic stimuli; overexpression studies have shown similar results with GSDMA [103] . Beyond these connections, in cells lacking pyroptosis via GSDMD-deficiency, caspase-1 can cleave caspase-3 and Bid to promote apoptotic cell death in response to inflammasome triggers such as LPS priming and poly(dA:dT) transfection, or during Salmonella infection [110, 111] . Furthermore, the APAF1-apoptosome has been shown to interact with caspase-11 when cells are challenged with bile acid; the result is caspase-3 cleavage and the execution of pyroptotic death in a GSDME-mediated process [112] . Other pore-forming molecules may also be involved in this crosstalk, as pannexin-1 activation downstream of caspase-8 or -9 activation leads to NLRP3 inflammasome formation in a GSDMD-and GSDME-independent process [113] . The depth and breadth of literature encompassing innate immune signaling and programmed cell death is impressive. Repeatedly, the literature acknowledges instances of crosstalk between components of pyroptosis, apoptosis, and necroptosis. This crosstalk occurs in context-dependent manners and is sometimes referred to as 'plasticity' or 'redundancy', with cell death components often labeled as 'molecular switches'. The overwhelming amount of evidence for the interconnectedness between cell death pathways has led to the conceptualization of PANoptosis as an inflammatory cell death pathway. The totality of biological effects in PANoptosis cannot be individually accounted for by pyroptosis, apoptosis, or necroptosis alone [2] [3] [4] [5] [6] [7] [8] [9] [10] [11] [12] [13] [14] [15] [16] [17] [18] [19] [20] . PANoptosis has been increasingly implicated in infectious and inflammatory diseases as well as in cancers and cancer therapies [2] [3] [4] [5] [7] [8] [9] [10] [11] [12] [13] [14] [15] [16] 18, 20, [114] [115] [116] [117] [118] . Here, we will focus on the most mechanistically wellcharacterized examples of PANoptosis and PANoptosomes [3, 7, [9] [10] [11] [12] 20] (Figure 1 ). As the conceptualization of PANoptosis implies, PANoptosis involves the activation of several molecules previously characterized as mediators of independent cell death pathways. For instance, Z-DNA-binding protein 1 (ZBP1) was previously known to induce necrosis in response to a mutant form of MCMV expressing a tetra-alanine RHIM substitution in vIRA (M45mutRHIM) [119] and was shown to interact with the necroptotic molecules RIPK1 and RIPK3 [120] , but more recent evidence has shown that ZBP1 acts as a cytosolic innate immune sensor for endogenous nucleic acids or during IAV infection to induce activation of the NLRP3 inflammasome, caspase-1, caspase-8, caspase-3, caspase-7 [7, 9, 11] , and MLKL [7, 11, 121] . Molecularly, ZBP1 mediates the formation of a multiprotein ZBP1-PANoptosome complex, containing ZBP1, RIPK3, RIPK1, caspase-8, caspase-6, ASC, and NLRP3 [10, 11] . To date, this complex has been characterized by immunoprecipitation [7, 10, 11] , and immunofluorescence has also shown colocalization of caspase-8 and RIPK3 with ASC specks in individual cells during IAV infection [122] . Further studies, including biochemical analyses and cryo-EM evaluation, are needed to fully understand how components come together in individual cells. The ZBP1-PANoptosome complex can also be implicated in tumorigenesis, where ADAR1 acts as a negative regulator to prevent the interaction between ZBP1 and RIPK3 and promote tumorigenesis. Limiting the interaction between ADAR1 and ZBP1 by sequestering ADAR1 in the nucleus, through treatment with nuclear transport inhibitors (KPT-330) in conjunction with IFN, potentiates PANoptosis and limits tumorigenesis [7] . Additionally, the inflammasome sensor AIM2 also initiates the formation of PANoptosome complexes during herpes simplex virus 1 (HSV1) and Francisella novicida infections. This PANoptosome, termed the AIM2-PANoptosome, contains AIM2, ZBP1, Pyrin, ASC, caspase-1, caspase-8, RIPK3, RIPK1, and FADD [12] . PANoptosomes have also been identified by immunoprecipitation during Yersinia infection, where RIPK1, RIPK3, caspase-8, FADD, ASC, and NLRP3 can be co-immunoprecipitated [3] . In the context of Yersinia infection, RIPK1 is necessary for activation of caspase-1, GSDMD, caspase-8, caspase-3, and caspase-7, but it negatively regulates the activation of MLKL, highlighting the multifaceted modulation of cell death effectors that can occur within PANoptosis [3] . PANoptosis has also been observed in response to TAK1 inhibition in macrophages, which can occur as a result of Yersinia infection due to its effector YopJ or in response to genetic mutations or treatment with TAK1 inhibitors [5, 20, 98, 100] . In the case of TAK1-deficient macrophages, spontaneous PANoptosis occurs and is characterized by the activation of the NLRP3 inflammasome, caspase-1, caspase-3, caspase-8, and MLKL [5, 20] ; stimulation with LPS in TAK1-deficient macrophages induces colocalization of RIPK1, ASC, and caspase-8 in a RIPK1 kinase-independent manner [20] . As with other cell death pathways, PANoptosis must be tightly regulated to control the execution of cell death. IRF1, a molecule long recognized for its roles in regulating cell death [123, 124] , is a key upstream regulator of PANoptosis. In the absence of IRF1 during IAV infection, ZBP1 protein expression, along with NLRP3 inflammasome, caspase-1, caspase-8, caspase-3, and MLKL activation, are all reduced [75] . In the context of colorectal tumorigenesis, IRF1 facilitates the activation of PANoptosis to limit tumorigenesis [8] . PANoptosis has also been observed to be driven by IRF1 in response to the combination of TNF and IFN-γ. TNF and IFN-γ release can occur physiologically during cytokine storm syndromes, including during SARS-CoV-2 infection [2] , and together they induce PANoptosis through the JAK/IRF1 signaling axis [2, 4] ; this observation has led to a mechanistic definition for cytokine storm as a life-threatening condition caused by excessive production of cytokines mediated by PANoptosis [125] . Additionally, the AIM2 inflammasome has also been shown to be regulated by IRF1 during Francisella infection [126] , suggesting a possible regulatory role of IRF1 in PANoptosis mediated by the AIM2-PANoptosome, although this remains to be investigated. Given the extensive history characterizing innate immune signaling and cell death molecules, complexes, and processes, it is important to revisit previous studies to potentially connect key concepts. The recent expansion of studies on PANoptosis further combined with a fresh look at the full body of literature in the cell death field suggests that the concept of PANoptosis has been hiding in plain sight (Tables 1 and 2 ). Many researchers have reported instances of cell death crosstalk, functional redundancy, or interconnectedness [127] , all of which sets the foundation for PANoptosis. As one key example, a wealth of literature exists on the apoptotic caspase-8 and how its loss or impaired functionality modulates multiple programmed cell death pathways and their plasticity in development and disease [62, [128] [129] [130] [131] [132] . Mice expressing a catalytically inactive version of caspase-8, i.e., Casp8 C362A/C362A , are embryonically lethal [130] . Cells carrying Casp8 C362A/C362A form ASC specks, induce activation of the inflammatory caspase substrate GSDMD, and activate apoptotic caspases, caspase-3 and caspase-7, to mediate cell death. While embryonic lethality in mice carrying Casp8 C362A/C362A can be partially rescued by deleting pyroptotic caspase-1 or ASC, mice with combined deletions of caspase-1/-11/RIPK3 have the best survival outcome [130] , suggesting that multiple cell death pathways are responsible for the embryonic lethality. Similar results are observed in mice carrying another enzymatically inactive caspase-8 mutation, Casp8 C362S/C362S , where lethality is rescued by combined deletion of MLKL/ASC or MLKL/caspase-1 [128] , suggesting that the apoptotic caspase-8 can play a scaffolding role, and multiple cell death effectors are involved in the cell death in mice carrying catalytically inactive caspase-8. On the one hand, mice carrying a non-cleavable version of caspase-8 (Casp8 Upstream, loss of ZBP1 is sufficient to prevent ileitis in both mice with caspase-8-and FADD-deficient IECs [131] . Collectively, these results align well with the PANoptosis concept, where there is a totality of biological effects in the phenotype that do not fit within pyroptosis, apoptosis, or necroptosis alone. [9] [10] [11] 133] ZBP1 [12, 134] AIM2 IRF1 [10, 30, 135, 136 ] Studies have consistently identified cell death crosstalk, plasticity, redundancy, interconnection, and molecular switches in evaluations of disease and cellular phenotypes. The totality of biological effects in these studies cannot be individually accounted for by pyroptosis, apoptosis, or necroptosis alone, leading to the conceptualization of PANoptosis. This table focuses on cell death in murine bone marrow-derived macrophages as a model. For genotypes representing a disruption in each programmed cell death pathway, the presence or absence of cell death (Cell death?) is denoted for each. , cell death occurs at levels similar to those seen in wild-type cells; D, decreased cell death as compared with wild-type; I, increased cell death as compared with wild-type; X, no cell death. [16, 18, 140, 141] Osteomyelitis [142] [143] [144] [145] [146] Dermatitis [147, 148] Dermatosis [62, [81] [82] [83] [84] 131] Embryonic lethality [130] Embryonic lethality [151, 152] Hypothermia genotype is considered as an apoptosis deletion only when the phenotype is the same as the Ripk3 −/− genotype, showing that the added deletion of apoptotic caspase-8 does not affect the phenotype. b Sharpin cpdm Casp8 −/− Ripk3 −/− mice could be born but did not survive to weaning. c Hoil −/− Casp8 −/− Ripk3 −/− mice succumb at late gestation through a process that appears to be independent of cell death, while Hoil −/− Casp8 +/− Ripk3 −/− mice undergo cell death-induced loss of yolk sac vascularization to contribute to lethality. Disease phenotypes in mouse models are associated with many different cell death molecules. Deletion of specific combinations can alleviate disease. The totality of biological effects in these studies cannot be individually accounted for by pyroptosis, apoptosis, or necroptosis alone, leading to the conceptualization of PANoptosis. For genotypes representing a disruption in each programmed cell death pathway, the presence or absence of disease (Disease?) is denoted for each. , disease or lethality occurs at levels similar to those seen in wild-type animals; D, decreased disease or lethality as compared with wild-type; X, no disease or lethality (rescued). Many other in vivo disease phenotypes can be assessed through the PANoptosis lens. For instance, the osteomyelitic disease observed in mice carrying a Pstpip2 cmo mutation is likely associated with PANoptosis. The bone disease in these mice is driven by aberrant production of IL-1β and is associated with inflammasome activation and cell death. Given the clear role of inflammasome-mediated cytokine release in the disease, it seemed likely that deletion of pyroptotic molecules, such as NLRP3 or caspase-1, would rescue the disease. However, this was not the case. Only mice lacking combinations of cell death molecules, including NLRP3/caspase-8/RIPK3 or caspase-1/-8/RIPK3, are protected from disease [16, 18] . Similar results are observed in the Sharpin cpdm mouse model, where mutations in SHARPIN, a linear ubiquitin chain assembly complex component critical for TNF signaling activation, result in skin inflammation. In these mice, deletion of NLRP3 or caspase-1 [142] or MLKL alone [143] delays, but does not prevent, the skin inflammation, while combined deletion of FADD/RIPK3 in epidermal cells rescues the inflammation [144] . Additionally, footpad inflammation in mice carrying the Ptpn6 spin mutation is not rescued by single deletions of caspase-1, NLRP3, RIPK3, MLKL, or the combined deletion of caspase-8/RIPK3 [147, 148] . The inflammatory disease in these mice, which resembles neutrophilic dermatosis in humans, is mediated by the RIPK1/IL-1α signaling axis, but combined deletion of caspase-8/RIPK3/RIPK1 is needed to prevent the inflammation [148] . Furthermore, innate immunity and programmed cell death have been connected to neurodegenerative diseases such as Alzheimer's disease (AD). Elevated levels of proinflammatory cytokines that can be released by cell death, such as TNF-α and IL-1β, are found in the brains and serum of people with AD and can cause neuroinflammation and AD-related pathologies [153] . The potential for PANoptosis is shown in studies with amyloid precursor protein/presenilin 1 (APP/PS1) transgenic mice carrying kinase-dead RIPK1 (Ripk1 D138N ). A deficiency in functional RIPK1 results in reduced disease and decreased proinflammatory IL-1β release [154] , linking necroptotic and pyroptotic molecules in this model. Given the multifaceted nature of the cell death in many in vivo disease phenotypes, it is likely that many others could also be considered within the PANoptosis concept. Several infection models have also demonstrated cell death crosstalk and interconnectivity that can be considered within the PANoptosis concept. As discussed above, ZBP1-mediated cell death during IAV infection is the prototypical example of PANoptosis. In initial studies, IAV-induced cell death was often viewed as redundant or thought to concurrently activate multiple cell death pathways [75, 121, 155] , but later work elucidated the molecular mechanisms of ZBP1-PANoptosome formation and established the complex [11] . ZBP1-mediated PANoptosis has also been implicated in tumorigenesis [7] and fungal infections with Candida albicans and Aspergillus fumigatus [14] . In response to other viruses, PANoptosis mediated through AIM2-PANoptosome formation has also been characterized during HSV1 infection [12] . Additionally, during hepatitis C virus infection, fluorescence microscopy has shown concurrent caspase-1 and caspase-3 activation [156] , suggesting simultaneous activation of these molecules in individual cells, which could be explained by PANoptosis. Bacterial pathogens have also been found to induce cell death where the molecular phenotypes observed do not fall within pyroptosis, apoptosis, or necroptosis activation alone, suggesting PANoptosis is occurring. In response to FlaTox, mice lacking either the combination of caspase-1/RIPK3 or the combination of caspase-8/RIPK3 still experience pathology, while combined loss of caspase-1/-8/RIPK3 provides protection [151] . Similarly, co-deletion of ASC or caspase-8/RIPK3 with caspase-1/-11 phenocopies NLRC4 deletion during challenge with flagellin or infection with Legionella [92, 151] . In Salmonella infection, 'flexible' cell death has been observed, and it has been reported that components of different cell death pathways interact. Salmonella-induced NLRC4 inflammasome activation drives cell death to clear bacteria from the host, and mice deficient in RIPK3 and caspase-1 are moderately impaired in their ability to clear the bacteria, while bacterial clearance in mice deficient in RIPK3, caspase-1, and caspase-8 is significantly more impaired [151] . Subsequent studies have shown that cells deficient in caspase-1/-11 or caspase-8/RIPK3 still undergo cell death in response to the infection, while cells deficient in caspase-1/-11/-8/RIPK3 [10] or cells deficient in caspase-1/-11/-12/-8/RIPK3 [135] are protected from cell death. Infection with Bacillus anthracis also activates multiple cell death effectors, including caspase-1, -8, and -3. Deletion of RIPK3 is not sufficient to protect cells from death during this infection, while the combined deletion of caspase-8/RIPK3 does prevent cell death [157] . Additionally, Pseudomonas aeruginosa induces cell death characterized by activation of caspase-1, GSDMD, caspase-8, -3, -7, and MLKL. While deletion of the pyroptotic sensors NLRP3 and NLRC4 together significantly reduces cell death in response to P. aeruginosa, combined deletion of caspase-1/-11/-8/RIPK3 provides full protection from cell death [137] . As each of these phenotypes cannot be explained by activation of pyroptosis, apoptosis, or necroptosis alone, they may fit in the category of PANoptosis. Having PANoptosis as a component of the innate immune response is likely advantageous on the organismal level. Inhibiting cell death effectors is a commonly used pathogen defense strategy [158] [159] [160] , and PANoptosis allows the cell to utilize multiple routes of cell death if one or more key cell death effectors is inhibited. Biologically, the redundancy displayed by PANoptosis ensures a fail-safe mechanism for cell death, thereby increasing the likelihood of organismal survival. During Yersinia infection, the bacteria secrete YopJ, a bacterial effector that inhibits TAK1-dependent inflammatory signaling [98, 100] . However, in response to the inhibition of TAK1, host cells undergo cell death characterized by activation of the NLRP3 inflammasome, caspase-1, -3, -7, and MLKL [5] . Recently, Yersinia infection was found to activate PANoptosis, with infection inducing the formation of a PANoptosome containing RIPK1, RIPK3, caspase-8, ASC, FADD, and NLRP3 [3] . Shigella flexneri also uses its effector molecules, OspC1 to inhibit caspase-8 and OspD3 to inhibit RIPK1/RIPK3, in an attempt to evade the cell death crosstalk [161] , which could give this bacterium a strategy to prevent PANoptosis. Experimental design often restricts how we test the innate immune response, limiting our ability to fully determine connections between molecular processes. The host response is likely more complex than we understand, but we can appreciate that the host must be able to recognize and respond to a variety of danger signals and cellular insults to execute the appropriate cellular response. It would likely be evolutionarily advantageous for a barrage of cellular insults to be neutralized using an integrated immune response, such as that stemming from PANoptosis. Therefore, it is important to be inclusive and consider the totality of biological effects in PANoptosis when studying cell death. These phenotypes may be missed when focusing on a single cell death pathway due to functional redundancies and overlaps between molecules. Additionally, the activation of molecular executioner signatures of pyroptosis, apoptosis, and necroptosis are not required simultaneously in an individual cell for a cell death process to fit within the PANoptosis concept. Furthermore, there are certainly conditions where pyroptosis, apoptosis, or necroptosis alone carries out the cell death, such as pyroptosis in wild-type cells exposed to LPS + ATP or necroptosis in wild-type cells exposed to TNF + zVAD. Only by studying the totality of the effects of programmed cell death in physiologically relevant models can we identify the important differences between these instances and PANoptosis. Multiple PANoptosome complexes have been associated with PANoptosis to date [3, 7, [9] [10] [11] [12] 20] , and additional work is required to fully understand these complexes. Their molecular composition may be flexible and dynamic. In the case of other cell deathinducing complexes, such as the NLRP3 inflammasome, there are canonical and noncanonical iterations; the same may also be true for PANoptosomes. Canonical PANoptosomes may contain caspase-8, RIPK3, and inflammasome components, as has been observed during IAV, HSV1, Francisella, and Yersinia infections [3, [10] [11] [12] , while non-canonical PANoptosomes may contain caspase-8 and RIPK3 as major molecules, as is likely the case during TNF + IFN-γ stimulation [2, 4] . More work is needed to continue to evaluate these complexes, the dynamics of their formation, and their regulation. Furthermore, in addition to the ZBP1-and AIM2-PANoptosome complexes that have been molecularly characterized based on their upstream sensors, it is likely that many other sensor-specific PANoptosomes exist and form in response to infections and conditions of altered cellular homeostasis that have not yet been tested. This has been found with inflammasomes and will likely be a conserved phenomenon. Whether sensors are strictly intracellular, or cell surface receptors are also involved, is also currently unknown. The complexity of tissue-and cell-specific responses, in relation to PANoptosis, should also be considered and further investigated. PANoptosis is a fluid process due to the many ways cell death can be executed. Some routes of cell death may be favored in certain tissues or cell types. Additionally, the PANoptotic response may be executed differently in various cell types based on differences in gene expression of key cell death molecules that exist among cells. It is also worth noting that many cancers are largely associated with dysregulation of apoptosis, further highlighting pathway specificity in some contexts. Understanding the subtle nuances which make each tissue and cell type unique may be useful in determining how PANoptosis occurs in order to harness this process for therapeutic purposes. Furthermore, there is a need to mechanistically understand the regulation of PANoptosis, as this could be the key to developing new clinical therapeutics for human cancers and diseases. IRF1 has been implicated as a central upstream regulator [2, 4, 8, 75] , and other components of this pathway should be elucidated. Inhibiting certain molecular components of cell death may enhance the activation of others, as has been observed previously [15, 137] , and this requires careful consideration during the drug design process. Perhaps targeting one aspect of cell death will result in subpar clinical therapies due to the redundancy of PANoptotic molecules. PANoptosis has been implicated across the disease spectrum, including in cerebral ischemia [114] ; bacterial, viral, and fungal infections [3, [9] [10] [11] [12] [13] [14] [15] , including oral infections [115, 116] ; inflammatory diseases [2, 5, 16, 18, 20] ; cancers [4, 7, 8] ; and cancer therapies [4, 7, 117, 118] . It will be important to improve our understanding of this pathway and identify how previous descriptions of crosstalk, plasticity, redundancies, molecular switches, and interconnectedness among cell death processes fit within this inclusive concept to gain a holistic picture of cell death. Only when we identify all the ingredients in the PAN can we begin to effectively target these molecules and develop novel therapeutics to save lives and improve patient outcomes. Acknowledgments: We apologize to our colleagues in the field whose work could not be cited owing to space limitations. We thank all the members of the Kanneganti laboratory for their comments and suggestions. We also thank XVIVO for assistance with scientific illustrations. The authors declare no conflict of interest. Master Regulators of NLRP3 Inflammasome/Pyroptosis, Apoptosis, and Necroptosis (PAN-optosis) Synergism of TNF-alpha and IFN-gamma Triggers Inflammatory Cell Death, Tissue Damage, and Mortality in SARS-CoV-2 Infection and Cytokine Shock Syndromes RIPK1 Distinctly Regulates Yersinia-Induced Inflammatory Cell Death, PANoptosis. Immunohorizons Inflammatory Cell Death, PANoptosis, Mediated by Cytokines in Diverse Cancer Lineages Inhibits Tumor Growth TAK1 restricts spontaneous NLRP3 activation and cell death to control myeloid proliferation Cutting edge: Proteolytic inactivation of poly(ADP-ribose) polymerase 1 by the Nlrp3 and Nlrc4 inflammasomes Interferon regulatory factor 1 regulates PANoptosis to prevent colorectal cancer ZBP1/DAI is an innate sensor of influenza virus triggering the NLRP3 inflammasome and programmed cell death pathways Identification of the PANoptosome: A Molecular Platform Triggering Pyroptosis Caspase-6 is a key regulator of innate immunity, inflammasome activation and host defense AIM2 forms a complex with pyrin and ZBP1 to drive PANoptosis and host defence The Zα2 domain of ZBP1 is a molecular switch regulating influenza-induced PANoptosis and perinatal lethality during development ZBP1 promotes fungiinduced inflammasome activation and pyroptosis, apoptosis, and necroptosis (PANoptosis) Impaired NLRP3 inflammasome activation/pyroptosis leads to robust inflammatory cell death via caspase-8/RIPK3 during coronavirus infection Dietary modulation of the microbiome affects autoinflammatory disease Targeted peptidecentric proteomics reveals caspase-7 as a substrate of the caspase-1 inflammasomes NLRP3 inflammasome plays a redundant role with caspase 8 to promote IL-1β-mediated osteomyelitis FADD and caspase-8 mediate priming and activation of the canonical and noncanonical Nlrp3 inflammasomes Innate immune priming in the absence of TAK1 drives RIPK1 kinase activity-independent pyroptosis, apoptosis, necroptosis, and inflammatory disease Caspases in Cell Death, Inflammation, and Pyroptosis Molecular mechanisms of cell death: Recommendations of the Nomenclature Committee on Cell Death Pro-inflammatory programmed cell death The inflammasome: A molecular platform triggering activation of inflammatory caspases and processing of proIL-beta Bacterial RNA and small antiviral compounds activate caspase-1 through cryopyrin/Nalp3 Cryopyrin activates the inflammasome in response to toxins and ATP Gout-associated uric acid crystals activate the NALP3 inflammasome Cytosolic flagellin requires Ipaf for activation of caspase-1 and interleukin 1beta in salmonella-infected macrophages Cytoplasmic flagellin activates caspase-1 and secretion of interleukin 1beta via Ipaf Differential activation of the inflammasome by caspase-1 adaptors ASC and Ipaf Innate immune sensing of bacterial modifications of Rho GTPases by the Pyrin inflammasome AIM2 activates the inflammasome and cell death in response to cytoplasmic DNA AIM2 recognizes cytosolic dsDNA and forms a caspase-1-activating inflammasome with ASC Prion-like polymerization underlies signal transduction in antiviral immune defense and inflammasome activation Unified polymerization mechanism for the assembly of ASC-dependent inflammasomes ASC, a novel 22-kDa protein, aggregates during apoptosis of human promyelocytic leukemia HL-60 cells Structure and assembly of the mouse ASC inflammasome by combined NMR spectroscopy and cryo-electron microscopy Immunological and inflammatory functions of the interleukin-1 family Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death Gasdermin D is an executor of pyroptosis and required for interleukin-1beta secretion GsdmD p30 elicited by caspase-11 during pyroptosis forms pores in membranes Pore-forming activity and structural autoinhibition of the gasdermin family Inflammasome-activated gasdermin D causes pyroptosis by forming membrane pores GSDMD membrane pore formation constitutes the mechanism of pyroptotic cell death Caspase-11 cleaves gasdermin D for non-canonical inflammasome signalling IRF2 transcriptionally induces GSDMD expression for pyroptosis A genome-wide screen identifies IRF2 as a key regulator of caspase-4 in human cells Apoptosis: A basic biological phenomenon with wide-ranging implications in tissue kinetics Apaf-1, a human protein homologous to C. elegans CED-4, participates in cytochrome c-dependent activation of caspase-3 Formation of apoptosome is initiated by cytochrome c-induced dATP hydrolysis and subsequent nucleotide exchange on Apaf-1 Cytochrome c and dATP-dependent formation of Apaf-1/caspase-9 complex initiates an apoptotic protease cascade Involvement of MACH, a novel MORT1/FADD-interacting protease, in Fas/APO-1-and TNF receptor-induced cell death FLICE, a novel FADD-homologous ICE/CED-3-like protease, is recruited to the CD95 (Fas/APO-1) death-inducing signaling complex Caspase cleaved BID targets mitochondria and is required for cytochrome c release, while BCL-XL prevents this release but not tumor necrosis factor-R1/Fas death Bid, a Bcl2 interacting protein, mediates cytochrome c release from mitochondria in response to activation of cell surface death receptors Cleavage of BID by caspase 8 mediates the mitochondrial damage in the Fas pathway of apoptosis tBID, a membrane-targeted death ligand, oligomerizes BAK to release cytochrome c Bid, Bax, and lipids cooperate to form supramolecular openings in the outer mitochondrial membrane The role of necroptosis in cancer biology and therapy Necroptosis in anti-viral inflammation Caspase-8 serves both apoptotic and nonapoptotic roles Cleavage of RIPK1 by caspase-8 is crucial for limiting apoptosis and necroptosis Caspase 8 inhibits programmed necrosis by processing CYLD Cleavage of RIP3 inactivates its caspase-independent apoptosis pathway by removal of kinase domain Initiation and execution mechanisms of necroptosis: An overview Molecular mechanisms of necroptosis: An ordered cellular explosion Mixed lineage kinase domain-like protein mediates necrosis signaling downstream of RIP3 kinase Mixed lineage kinase domain-like is a key receptor interacting protein 3 downstream component of TNF-induced necrosis Ubiquitylation of MLKL at lysine 219 positively regulates necroptosis-induced tissue injury and pathogen clearance Structural mechanism for NEK7-licensed activation of NLRP3 inflammasome NLRP3 activation and mitosis are mutually exclusive events coordinated by NEK7, a new inflammasome component A Genome-wide CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats) Screen Identifies NEK7 as an Essential Component of NLRP3 Inflammasome Activation NEK7 is an essential mediator of NLRP3 activation downstream of potassium efflux as a live-or-die checkpoint in stressed cells by regulating NLRP3 inflammasome IRF1 Is a Transcriptional Regulator of ZBP1 Promoting NLRP3 Inflammasome Activation and Cell Death during Influenza Virus Infection IRF8 Regulates Transcription of Naips for NLRC4 Inflammasome Activation NINJ1 mediates plasma membrane rupture during lytic cell death Toward targeting inflammasomes: Insights into their regulation and activation pyroptosis and apoptosis: An intricate game of cell death Fas triggers an alternative, caspase-8-independent cell death pathway using the kinase RIP as effector molecule Catalytic activity of the caspase-8-FLIP(L) complex inhibits RIPK3-dependent necrosis RIP3 mediates the embryonic lethality of caspase-8-deficient mice The Pseudokinase MLKL and the Kinase RIPK3 Have Distinct Roles in Autoimmune Disease Caused by Loss of Death-Receptor-Induced Apoptosis Targeted disruption of the mouse Caspase 8 gene ablates cell death induction by the TNF receptors, Fas/Apo1, and DR3 and is lethal prenatally Mutations that prevent caspase cleavage of RIPK1 cause autoinflammatory disease Pleiotropic defects in lymphocyte activation caused by caspase-8 mutations lead to human immunodeficiency Caspase-1 activity is required to bypass macrophage apoptosis upon Salmonella infection Inflammasome activation leads to Caspase-1-dependent mitochondrial damage and block of mitophagy AIM2/ASC triggers caspase-8-dependent apoptosis in Francisella-infected caspase-1-deficient macrophages AIM2 and NLRP3 inflammasomes activate both apoptotic and pyroptotic death pathways via ASC Salmonella infection induces recruitment of Caspase-8 to the inflammasome to modulate IL-1β production Inhibition of caspase-1 or gasdermin-D enable caspase-8 activation in the Naip5/NLRC4/ASC inflammasome Caspase-1 Engagement and TLR-Induced c-FLIP Expression Suppress ASC/Caspase-8-Dependent Apoptosis by Inflammasome Sensors NLRP1b and NLRC4 Inflammasome activation causes dual recruitment of NLRC4 and NLRP3 to the same macromolecular complex Caspase-8 scaffolding function and MLKL regulate NLRP3 inflammasome activation downstream of TLR3 Active MLKL triggers the NLRP3 inflammasome in a cell-intrinsic manner MLKL Activation Triggers NLRP3-Mediated Processing and Release of IL-1β Independently of Gasdermin-D Caspase-8 induces cleavage of gasdermin D to elicit pyroptosis during Yersinia infection Caspase-8-dependent gasdermin D cleavage promotes antimicrobial defense but confers susceptibility to TNF-induced lethality Pathogen blockade of TAK1 triggers caspase-8-dependent cleavage of gasdermin D and cell death Pyroptosis and apoptosis pathways engage in bidirectional crosstalk in monocytes and macrophages Shiga Toxin/Lipopolysaccharide Activates Caspase-4 and Gasdermin D to Trigger Mitochondrial Reactive Oxygen Species Upstream of the NLRP3 Inflammasome Gasdermin pores permeabilize mitochondria to augment caspase-3 activation during apoptosis and inflammasome activation Single-cell analysis of pyroptosis dynamics reveals conserved GSDMD-mediated subcellular events that precede plasma membrane rupture The DFNA5 gene, responsible for hearing loss and involved in cancer, encodes a novel apoptosis-inducing protein Chemotherapy drugs induce pyroptosis through caspase-3 cleavage of a gasdermin Cleavage of DFNA5 by caspase-3 during apoptosis mediates progression to secondary necrotic/pyroptotic cell death Gasdermin E permits interleukin-1 beta release in distinct sublytic and pyroptotic phases Caspase-1 initiates apoptosis in the absence of gasdermin D Caspase-1 cleaves Bid to release mitochondrial SMAC and drive secondary necrosis in the absence of GSDMD Apaf-1 Pyroptosome Senses Mitochondrial Permeability Transition Extrinsic and intrinsic apoptosis activate pannexin-1 to drive NLRP3 inflammasome assembly Do pyroptosis, apoptosis, and necroptosis (PANoptosis) exist in cerebral ischemia? Evidence from cell and rodent studies PANoptosis: A New Insight Into Oral Infectious Diseases Real-Time Induction of Macrophage Apoptosis, Pyroptosis, and Necroptosis by Enterococcus faecalis OG1RF and Two Root Canal Isolated Strains Phosphorylated NFS1 weakens oxaliplatin-based chemosensitivity of colorectal cancer by preventing PANoptosis Self-assembled polymeric nanocarrier-mediated co-delivery of metformin and doxorubicin for melanoma therapy /ZBP1/DLM-1 complexes with RIP3 to mediate virus-induced programmed necrosis that is targeted by murine cytomegalovirus vIRA Receptor-interacting protein homotypic interaction motif-dependent control of NF-kappa B activation via the DNA-dependent activator of IFN regulatory factors DAI senses influenza A virus genomic RNA and activates RIPK3-dependent cell death From pyroptosis, apoptosis and necroptosis to PANoptosis: A mechanistic compendium of programmed cell death pathways An IRF-1-dependent pathway of DNA damage-induced apoptosis in mitogen-activated T lymphocytes Cellular commitment to oncogene-induced transformation or apoptosis is dependent on the transcription factor IRF-1 The 'cytokine storm': Molecular mechanisms and therapeutic prospects The transcription factor IRF1 and guanylate-binding proteins target activation of the AIM2 inflammasome by Francisella infection Molecular crosstalk between apoptosis, necroptosis, and survival signaling Caspase-8 is the molecular switch for apoptosis, necroptosis and pyroptosis Caspase-8-Dependent Inflammatory Responses Are Controlled by Its Adaptor, FADD, and Necroptosis Activity of caspase-8 determines plasticity between cell death pathways FADD and Caspase-8 Regulate Gut Homeostasis and Inflammation by Controlling MLKL-and GSDMD-Mediated Death of Intestinal Epithelial Cells Mutation of a self-processing site in caspase-8 compromises its apoptotic but not its nonapoptotic functions in bacterial artificial chromosome-transgenic mice Cutting Edge: Caspase-8 Is a Linchpin in Caspase-3 and Gasdermin D Activation to Control Cell Death, Cytokine Release, and Host Defense during Influenza A Virus Infection Francisella infection triggers activation of the AIM2 inflammasome in murine dendritic cells Flexible Usage and Interconnectivity of Diverse Cell Death Pathways Protect against Intracellular Infection Caspase-11 increases susceptibility to Salmonella infection in the absence of caspase-1 NLRC4 Deficiency Leads to Enhanced Phosphorylation of MLKL and Necroptosis Immune recognition of Pseudomonas aeruginosa mediated by the IPAF/NLRC4 inflammasome Pseudomonas aeruginosa activates caspase 1 through Ipaf A missense mutation in pstpip2 is associated with the murine autoinflammatory disorder chronic multifocal osteomyelitis Inflammasome-independent IL-1β mediates autoinflammatory disease in Pstpip2-deficient mice The Inflammatory Caspases-1 and -11 Mediate the Pathogenesis of Dermatitis in Sharpin-Deficient Mice TNFR1-dependent cell death drives inflammation in Sharpin-deficient mice Sharpin prevents skin inflammation by inhibiting TNFR1-induced keratinocyte apoptosis A spontaneous mutation characterized by chronic proliferative dermatitis in C57BL mice Cutting Edge: RIP1 kinase activity is dispensable for normal development but is a key regulator of inflammation in SHARPIN-deficient mice RIP1-driven autoinflammation targets IL-1alpha independently of inflammasomes and RIP3 Tyrosine Kinase SYK Licenses MyD88 Adaptor Protein to Instigate IL-1alpha-Mediated Inflammatory Disease LUBAC is essential for embryogenesis by preventing cell death and enabling haematopoiesis Caspase-8 Collaborates with Caspase-11 to Drive Tissue Damage and Execution of Endotoxic Shock NAIP-NLRC4 Inflammasomes Coordinate Intestinal Epithelial Cell Expulsion with Eicosanoid and IL-18 Release via Activation of Caspase-1 and -8 Rapid induction of inflammatory lipid mediators by the inflammasome in vivo The role of neuroimmunomodulation in Alzheimer's disease RIPK1 mediates a disease-associated microglial response in Alzheimer's disease RIPK3 activates parallel pathways of MLKL-criven necroptosis and FADD-mediated apoptosis to protect against Influenza A virus Crosstalk Between Pyroptosis and Apoptosis in Hepatitis C Virus-induced Cell Death. Front Bacillus anthracis induces NLRP3 inflammasome activation and caspase-8-mediated apoptosis of macrophages to promote lethal anthrax Viral modulation of the host response via crmA/SPI-2 expression Manipulation of Host Cell Death Pathways by Herpes Simplex Virus Structure-function analysis of the interaction between Bax and the cytomegalovirus-encoded protein vMIA A unique bacterial tactic to circumvent the cell death crosstalk induced by blockade of caspase-8