Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Apr 21.
Published in final edited form as: Biochim Biophys Acta. 2011 Jun 16;1824(1):113–122. doi: 10.1016/j.bbapap.2011.06.005

Proliferative versus Apoptotic Functions of Caspase-8 Hetero or Homo: The Caspase-8 Dimer Controls Cell Fate

Bram J van Raam 1,*, Guy S Salvesen 1
PMCID: PMC3993904  NIHMSID: NIHMS566800  PMID: 21704196

Abstract

Caspase-8, the initiator of extrinsically-triggered apoptosis, also has important functions in cellular activation and differentiation downstream of a variety of cell surface receptors. It has become increasingly clear that the heterodimer of caspase-8 with the long isoform of cellular FLIP (FLIPL) fulfills these pro-survival functions of caspase-8. FLIPL, a catalytically defective caspase-8 paralog, can interact with caspase-8 to activate its catalytic function. The caspase-8/FLIPL heterodimer has a restricted substrate repertoire and does not induce apoptosis. In essence, caspase-8 heterodimerized with FLIPL prevents the receptor interacting kinases RIPK1 and -3 from executing the form of cell death known as necroptosis. This review discusses the latest insights in caspase-8 homo- vs. heterodimerization and the implication this has for cellular death or survival.

Keywords: Apoptosis, Caspase, FLIP, Necroptosis, Receptor Interacting Protein Kinase

1 Introduction

1.1 Cell death and caspases

Caspases are cysteine proteases and the first member of this family was discovered some 20 years ago as the protease responsible for the maturation of interleukin-1β [1,2]. Initially, this enzyme was called Interleukin Conversion Enzyme (ICE) but is now referred to as caspase-1. A related cysteine protease was discovered a few months later in the nematode worm Caenorhabditis elegans and called CED3 [3,4]. In C. elegans, CED3 was found to be responsible for the execution of developmental cell death and ever since, caspases have been almost synonymous with cell death [5]. However, it was soon recognized that in mammals at least two distinct branches of the caspase family have evolved with distinct functions; one branch involved in the initiation and execution of programmed cell death and one in the regulation of inflammation [6]. Caspases cleave their substrates exclusively after an aspartic acid residue (Asp/D). For a detailed overview of the activation mechanisms and substrate preferences of the caspases, we refer the reader to previous reviews [6,7]. Caspase-dependent programmed cell death is generally referred to as apoptosis, although caspase-1 is implicated in the induction of an inflammation-associated form of programmed cell death known as pyroptosis [8]. Two major pathways leading to apoptosis have been described, ‘intrinsic’ and ‘extrinsic’. Intrinsic apoptosis is initiated by caspase-9 which, for its activation, depends on the release of cytochrome c and other pro-apoptotic factors from the mitochondria through a permeability transition pore formed by the pro-apoptotic members of the Bcl-2 family, primarily Bid and Bax. Pro-survival members of the Bcl-2 family counteract the formation of this pore and thus the release of apoptogenic proteins from the mitochondria, as reviewed in detail by Lindsay et al. [9]. Extrinsic apoptosis occurs downstream of death receptor signaling and is initiated by caspases-8 and -10, as will be discussed in greater detail below. Since Bid is a target of caspase-8/10 activity, the two pathways have been proposed to be inter-linked, although probably only in certain cells or tissues [10]. In the absence of caspase activation, another form of programmed cell death, now generally referred to as ‘necroptosis’, can be triggered [11,12]. Activation of this pathway is actively opposed by caspases, primarily by caspase-8. The objective of this review is to discuss the recent insights in the pro-survival role of caspase-8 that is linked to its catalytic function, primarily its role in T-cell activation and homeostasis by control of necroptotic death and the activation mechanism of caspase-8 by the inactive caspase homolog FLIPL.

1.2 All the colors of the caspases

As mentioned above, human caspases seem to have evolved to be either involved in the regulation of apoptosis or inflammation. In Figure 1A, the pro-apoptotic members of the caspase family are marked in green, while the pro-inflammatory members are marked in red. Caspase-2 and -14, which have neither clear apoptotic nor clear inflammatory functions, are marked in black. The pro-apoptotic members of the caspase family are subdivided in the initiators of apoptosis (caspases-8, -9 and -10 in humans) and the executioners of apoptosis (caspase-3, -6 and -7). The difference between the two is that the initiators have a relatively large N-terminal dimerization domain, either a Death Effector Domain (DED; caspases-8 and -10) or a structurally related Caspase Recruitment Domain (CARD; caspase-9) and are expressed as monomers that require dimerization for activation. In contrast, the executioners only have short N-terminal pro-domains, exist as inactive dimeric zymogens in solution and require limited proteolytic cleavage between the large and small subunits for activation [6]. A general overview of the domain organization of the caspases is shown in Figure 1B. Caspase-2, which also has a CARD, is sometimes grouped among the initiators of apoptosis, however its function is hotly debated and it may either be involved in the DNA damage response, inducing either DNA repair or DNA-damage induced apoptosis, heat-shock induced apoptosis, the release of cytochrome c from the mitochondria [13-19] or none of the above. Another ‘orphan’ of the caspase family is caspase-14, which is exclusively expressed in keratinocytes and involved in their differentiation [20]. The most-studied member of the pro-inflammatory caspases is caspase-1, which is essential for the maturation of the pro-inflammatory cytokine interleukin-1β (IL-1β) as mentioned before [5]. The exact functions of the related caspases- 4 and -5 remain unknown, although they have been implicated in a number of processes [21-23]. Caspase-12, finally, is only expressed in a truncated, catalytically inactive form in most humans, with the exception of a small percentage of the sub-Saharan African population that expresses the full-length enzyme. It is thought that full-length caspase-12 may have a protective effect against certain infections in neonates [24-26]. Alternatively, caspase-12 may acts as a dominant negative in the regulation of caspase-1 activity [27]. Of late, it has become increasingly clear that the pro-apoptotic caspases, and in particular caspase-8, also have important functions in survival and development.

Figure 1. The human caspases.

Figure 1

Figure 1

A: Phylogenetic tree of all the human caspases. Pro-apoptotic caspases are marked in green, pro-inflammatory caspases in red. Caspases with no clear apoptotic of inflammatory function are marked in black. Image generated with Genious Tree Builder using the full length protein sequence of the human caspases using Genious Pro v5.1.6, as developed by Biomatters Ltd.

B: Over all organization of the initiator and executioner caspases. The yellow circle (●) on the ‘large’ subunit denotes the active site cysteine; blue circles (●) in the linkers denote caspase cleavage sites.

2 The developmental role of caspase-8

2.1 Caspase-8 knockout mice

It is the human condition to classify, subdivide and order. However, nature as such is not always as organized as we would like it to be. Therefore, when the first knock-out mice for apoptotic caspases were generated, it came as a bit of surprise that their phenotypes were not very apoptotic. The main defects of the caspase-3 and -9 knockout mice appeared to be in brain development, whereas apoptosis of hepatocytes or lymphocytes was not very much affected. For an extensive review of caspase knockout mice, we refer the interested reader to Los et al. [28]. The gene for caspase-10 was lost in the ancestor of the rodent lineage [29] – see Figure 2, but the caspase-8 knockout has the most dramatic phenotype of all caspase knockouts as these knockout mice die in utero at around day 11 of gestation with primary defects in cardiac development, growth retardation and hematopoietic progenitor deficiency [30]. Importantly, knockout of two other members of the complex in which caspase-8 is activated -FADD and FLIPL- both revealed a very similar phenotype [31,32]. This indicates that formation of this complex, the Death Inducing Signaling Complex (DISC) and caspase-8 activation does not only trigger apoptosis but is also essential for embryonic development. Further studies on mice with tissue specific knockout of caspase-8 revealed an essential role for this caspase in the development of a variety of tissues, most notably the endothelial cells of the heart and vascular system, lymphocytes and monocytes. Only in the liver, it appeared, the apoptotic functions of caspase-8 were primarily affected [33]. Whereas catalytically active caspase-8 is required to rescue the lymphocyte development in caspase-8 deficient mice [34], the caspase-8 DEDs alone appear to be required to rescue the terminal differentiation defect of endothelial, epithelial and myeloid cells [35]. Furthermore, mice deficient in caspase-8 in basal epidermal keratinocytes suffer from chronic skin inflammation, as an apparent consequence of IRF3 hyper-activation [36]. This phenotype, however, is not rescued by catalytically inactive caspase-8. How expression of the caspase-8 DEDs alone can be sufficient to induce terminal differentiation in certain cells, is as yet unclear, but implies an important non-catalytic role for caspase-8 [37,38].

Figure 2. Schematic of the caspase-8 phylogeny.

Figure 2

Two distinct caspase-8 family members arose in the chordate branch of the eukaryotes. Caspase-18 and the ancestor of -8 and -10 we call ‘caspase-810’ in this schematic, are still found in fishes. Later on in evolution, caspase-8 and -10 branched off from caspase-810. Birds and lizards express three apical caspases in the DR pathway; caspase-8, -10 and -18. Mammals subsequently lost caspase-18, while rodents lost caspase-10. See also Eckhart et al. [158].

2.2 Caspase-8/10 deficiencies in humans

In humans, caspase-8 deficiency is compatible with development, as demonstrated by the case of a family with a detrimental mutation in the caspase-8 gene [39]. In homozygous individuals, lymphocyte apoptosis and activation is impaired, but overall development is normal. Besides caspase-8, humans, contrary to mice, also express the highly homologous caspase-10, which has similar substrate specificity to caspase-8 [29] and at least some overlapping functions, but can not completely substitute for loss of caspase-8 [40-42]. While mutations or deficiencies in caspase-10 are associated with autoimmune lymphoproliferative syndrome (ALPS) [43,44], expression of either caspase-8 or caspase-10 is apparently sufficient for normal human development, but both are required to execute the apoptotic functions, at least in lymphocytes. It should also be noted that at least one mutation in caspase-10 that was originally associated with ALPS, V410I, is actually a common polymorphism in the healthy Danish population [45].

3 Activation of caspase-8/10

3.1 Dimerization vs. cleavage

In general, caspases can only be active as dimers, since neither the active site dyad nor the substrate pocket can be formed in the monomeric form [6]. In fact, it can not be stressed enough that dimerization is an absolute requirement for caspase activation. When caspase cleavage is detected, for example by Western blot, this does not signify that the caspase in question has been activated; merely that it has been cleaved. Active executioner caspases maintain the same core structure as zymogens, but undergo substantial re-arrangement of crucial surface loops after the intersubunit linker has been cleaved, leading to formation of the active site. Initiator caspases, on the other hand, have to undergo dimerization and sub sequent activation by induced proximity in large multi-protein complexes [46-48]. Caspase-8 and -10 alike are activated at the DISC, downstream of death-receptor signaling [46]. Upon ligation, death-receptors such as the tumor-necrosis factor receptor (TNFR) or Fas, multimerize in the cell membrane which leads to a conformational change in the intracellular domain of the receptor [49,50]. This, in turn, leads to recruitment and multimerization of the adaptor protein FADD, which contains both a Death Domain (DD) that interacts with the receptor, and a DED that recruits caspase-8/10. At this point, we realize that many authors state in their papers that caspases-8 and -10 are activated by oligomerization. This, however, is incorrect. The DISC is a high order oligomeric ensemble, and provides a platform for dimerization of caspases-8 and 10. Thus, caspases-8 and -10 are recruited to the DISC as intact monomers [51] and recruitment of the caspases to the DISC subsequently leads to their dimerization and activation through induced proximity [29,48,52]. Although the caspase-8 dimer is active in the absence of proteolytic cleavage within the intersubunit linker, cleavage stabilizes the dimer and leads to a caspase species with increased activity [53-59]. Finally, caspase-8 also cleaves itself between the DEDs and the large subunit, resulting in release of the stable dimer from the DISC [60]. Cleavage in the absence of dimerization does not lead to activation, but cleavage after dimerization increases both the activity and the stability of the dimer [54,56]. However, it has been suggested in several cases that caspase-8 can be activated through cleavage by another protease. The serine protease granzyme B, which is known to activate both caspase-3 and -7 during cytotoxic T-cell induced death, was shown to cleave caspase-8 as well [61]. In another case, caspase-6 was shown to cleave caspase-8 between the large and small subunit which was associated with Bid-dependent cytochrome c release from isolated mitochondria in a cell free system [62]. In this latter study, it was not excluded that recombinant caspase-6 can also cleave Bid directly at high concentrations or that caspase-6-cleaved caspase-8 has a decreased threshold for self-dimerization, possibly even in ‘mitosomes’ on the mitochondrial membrane, as has been suggested in other studies [63-65]. Finally, it has been suggested that the lysosomal protease cathepsin D can cleave and activate caspase-8 during neutrophil apoptosis [66]. In all of these cases, dimerization is most likely a prerequisite for activation whereas cleavage stabilizes the dimers and increases their activity.

3.2 Activation by FLIPL

A homologue of caspase-8, referred to as cellular Flice Inhibitory Protein (cFLIP) can also be recruited to the DISC. This homolog contains inactivating mutations in the catalytic site and is therefore catalytically dead. Heterodimerization with the long isoform of cFLIP (FLIPL) leads equally to activation of caspase-8/10 [67-70]. The unprocessed heterodimer has a restricted substrate repertoire and is no longer capable of cleaving and activating the main pro-apoptotic targets downstream of caspase-8/10 activation, such as caspase-3 and Bid, as recently demonstrated in two independent studies utilizing in vitro generated caspase-8/FLIPL heterodimers [57,71]. FLIPL was simultaneously identified by several groups, and data presented in these publications were highly controversial regarding whether FLIPL was pro- or anti-apoptotic. Some thought FLIPL to be an inhibitor of caspase-8, hence the name, and others thought it to be an independent caspase-8-like cell death inducer [72-76]. This controversy was settled when it was recognized that FLIPL only prevented cell death upon over-expression by occupying all the binding sites for caspase-8 at the DISC [77]. It now appears that caspase-8 has a wider array of functions besides inducing cell death and it appears to be the heterodimer, rather than the homodimer, which is involved in executing the non-death functions of caspase-8 [57,71,78]. Furthermore, the developmental phenotype observed in the caspase-8 knockout mouse is shared with both the FLIPL knockout and the FADD knockout [31,32], and FLIPL has been shown to be essential for cellular differentiation and activation in a number of studies [68,78-86]. Altogether, this indicates that it is FLIPL-activated caspase-8 that fulfills the non-apoptotic functions of caspase-8 prior to autoprocessing, rather than the caspase-8 homodimer - see Figure 3 for a schematic of homo- versus heterodimerization of caspase-8. Interestingly, whereas the uncleaved caspase-8 heterodimer with FLIPL can no longer cleave Bid and induce intrinsic apoptosis, the heterodimer of caspase-10 with FLIPL still can [29,57]. This implies that at least in that respect, caspases-8 and -10 are functionally different, but in mice, who do not express caspase-10, these functions are apparently subsumed by caspase-8. It should be noted that whereas human caspase-8 is cleaved twice in the intersubunit linker, mouse caspase-8 only has one cleavage site. The caspase-8/FLIPL heterodimer can process the first site with some difficulty, after which it can process the second site to become fully active. Since murine caspase-8 does not contain this first site, it is still an open question if and when murine caspase-8 dimerized with FLIPL can be activated to form an apoptotic species or whether in mice a secondary event is required to fully activate caspase-8 in the heterodimer.

Figure 3. Caspase-8 activation through homo- vs. heterodimerization.

Figure 3

Caspase-8 (green) can either homodimerize with another molecule of caspase-8, leading to a homodimer wherein caspase-8 is fully processed and induces apoptosis (top) or heterodimerize with FLIPL (blue) to form a heterodimer wherein FLIPL is primarily processed to induce cell survival (bottom). In either case, dimerization is mediated by the adaptor protein FADD (violet).

3.3 Structural insights into caspase-8 activation

Several crystal structures of the fully processed inhibitor-bound caspase-8 dimer have been published [87-91]. However, these structures allow little insight into the activation mechanism of caspase-8. The structure of monomeric un-processed caspase-8 as obtained by nuclear magnetic resonance (NMR) spectroscopy, was described only recently by Keller et al. [53]. Caspase-8 readily dimerizes and auto-processes during expression in E. coli and the unprocessed monomeric form is therefore hard to obtain in sufficient amounts to allow structural studies. The monomeric form of caspase-8 has a typical caspase fold, consisting of a six-stranded β sheet formed by five parallel and one anti-parallel β strand. This β sheet is flanked by a total of five α helices, two on one side of the structure and three on the other. Protruding from these structured regions are four loops and a short linker. The first of these loops, the one that occupies the active site in monomeric caspases, contains a short α helix and upon dimerization and cleavage of caspase-8, this loop rotates along a perpendicular axis by ~90° to allow formation of the active site, as illustrated in Figure 4A. Even after the first loop has been re-arranged and the active site has formed in the dimer, the linker between the large and small subunits occupies the active site in the structure, hindering access to the active site cysteine (the red helix in Figure 4A). Keller et al. suggested that processing of the linker, possibly through autocatalysis, opened the active site and allowed full activation of caspase-8. However, even a mutant that could not be cleaved in either of the two cleavage sites in the linker generated some activity upon dimerization while a mutant incapable of dimer formation but with a cleavable linker remained completely inactive, stressing once more the importance of dimer formation [53,54]. The only way to generate robust activity from a unprocessed caspase, is to heterodimerize it with FLIPL, as demonstrated by Pop et al.[57]. Interestingly, Boatright et al. [67], and later Yu et al.[70], showed that zymogen caspase-8 forms a much tighter interaction with FLIPL than with another molecule of caspase-8. As many as 8 hydrogen bonds are formed in the dimer interface between FLIPL and caspase-8, as compared to only 4 bonds in the caspase-8 homodimer [70]. The active site and the gross overall structural conformation of caspase-8 in this complex is very similar to active site of the homodimer, as illustrated in Figure 4, and therefore the restricted substrate specificity of the unprocessed heterodimer is likely caused by as yet undiscovered principles rather than a structurally different substrate cleft [57]. Since FLIPL is catalytically inactive and can therefore not process its binding partner, the caspase-8/FLIPL heterodimer is likely to remain unprocessed in cells until it is cleaved by an external protease, e.g. caspase-6. Within the caspase-8/FLIPL heterodimer, caspase-8 prefers to process FLIPL over itself and processing of free FLIPL has been shown to markedly increase the recruitment of caspase-8 [70]. After cleavage, FLIPL recruits other components to the complex with caspase-8, such as TRAF2, RIPK1 and NEMO, resulting in NF-κB activation [79,80,82]. Therefore, the heterodimer of caspase-8/FLIPL containing unprocessed caspase-8 signals towards cellular survival and activation, rather than towards apoptosis, and it is only after the caspase-8 subunit of the heterodimer has been processed that this caspase species becomes pro-apoptotic.

Figure 4. Structural insights in caspase-8 activation.

Figure 4

A: Structural overlay of the caspase-8 monomeric zymogen (green) and the substrate bound, fully-processed, caspase-8 dimer (orange; only one caspase-8 subunit is shown). During dimerization, a loop containing a small helix (in red) translocates from the active site (1), as indicated by the red arrow. Afterwards, the linker (blue) between the large and small subunits gets processed (2), opening up the active site completely for substrate binding. The inhibitor Z-EVD-CMK, in yellow, indicates the location of the active site cleft in the structure.

B: Structural overlay of the caspase-8 homo-dimer (earth colors) versus the caspase-8/FLIPL heterodimer (blues). Over all structural changes upon formation of either the homodimer or the heterodimer are grossly similar.

C-E: Comparison of the substrate cleft in the monomer (C) versus the peptide-bound homodimer (D) and the peptide-bound heterodimer (E). The substrate cleft is closed in the monomeric zymogen, whereas the cleft is accessible for substrate binding in both dimers. The synthetic peptide Ac-IETD-CHO is shown in magenta bound in the substrate cleft of the heterodimer (E). Based on PDB entries 1QDU, 2K7Z and 3H11 [53,70,88]. Images generated with PyMOL v1.4.

4. Programmed Necrosis

4.1 Necroptosis; death in the absence of caspase activation

By definition, forms of cell death that occur in the absence of caspases activation cannot be apoptosis, [92]. It was first observed in the murine fibroblastoma cell line L929 that if the cells were stimulated with TNFα in the presence of a broad spectrum caspases inhibitor the cells would die with a necrotic morphology [93]. This form of cell death was associated with high mitochondrial reactive oxygen species (ROS) production and loss of plasma membrane integrity [93,94], and was later recognized to occur in other cells as well upon stimulation with TNFα in the absence of caspase activation, as reviewed by Denecker et al. [95]. Further studies, performed by introducing the cowpox virus serpin and caspase-8 inhibitor CrmA [96] in the cells, revealed that in particular inhibition of caspase-8 triggered this form of cell death, now generally known as ‘necroptosis’ [93,97,98]. The serine/threonine kinase RIPK1 was discovered to be the initiator of necroptotic death in a screen for inhibitors of this form of cell death [11] and, more recently, three independent groups identified RIPK3 as a downstream effector of RIPK1 after extensive siRNA screening [99-101]. RIPK3 is thought to induce a switch in the cell’s metabolism, leading to the increase of mitochondrial ROS production that culminates in cell death [101,102]. Altogether, this strongly implies that necroptosis is an alternative form of programmed cell death, rather than an accidental form of death as once thought. Caspase-8 has been shown to cleave, and presumably inactivate, both RIPK1 and RIPK3 and thus acts as a negative regulator of this pathway on at least two different levels [103-106]. It should be noted that RIPK1 was initially identified as an activator of the NF-κB pathway of survival downstream of Fas/CD95 and TRAIL-receptor (DR4/5) signaling, but has later been suggested to be non-essential for activating NF-κB downstream of TNFR1 signaling [105,107,108]. However, Jurkat cells deficient in RIPK1 fail to activate NF-κB downstream of TNFR1, although it should be noted that these cells were selected for their inability to activate NF-κB and it cannot be excluded that they lack one or more other components in the NF-κB signaling cascade [109]. The main difference between the two death receptor signaling pathways appears to be that the TNFR1 receptor primarily signals through the adaptor TRADD and only secondarily through FADD [110], whereas DR4/5, similar to CD95/Fas primarily recruits the adaptor FADD [46]. The latter is disposable for activating RIPK1 as such, since FADD deficient T-cells or cells expressing dominant negative FADD are very susceptible to RIPK1-dependent necroptosis [111,112]. However, FADD is essential for the formation of both homo- and heterodimers of caspase-8. Thus, FLIPL-dependent NF-κB activation depends on FADD and, apparently, also on RIPK1. The main function of TRADD in the complex may be to attract the ubiquitin ligase TRAF2 which, through ubiquitination of RIPK1, may alter the outcome of DR signaling [113]. RIPK1 undergoes extensive ubiquitination on a single conserved lysine residue and whether it signals life or death appears to depend on whether the ubiquitin chain formed is K48, K63, K11-linked or linear [102,114-120]. A growing number of ubiquitin ligases and de-ubiquitinating enzymes regulate these functions of RIPK1, but discussing these is beyond the scope of this review. An excellent review on RIPK1 regulation by ubiquitination was recently published by Vandenabeele et al. [121]. However, novel regulators of the pathway are currently published on a regular basis.

4.2 Necroptosis versus Apoptosis

As mentioned before, both RIPK1 and RIPK3 have been identified as caspase-8 substrates [103,105,106]. Cleavage of RIPK1 prevents it from inducing necroptosis, while its C-terminal DD appears to be involved in DISC formation and can thus initiate apoptosis [95,122]. In both T-cell activation and macrophage development, caspase-dependent cleavage of RIPK1 has been shown to be an essential step [104,123-125]. In the absence of RIPK1 cleavage, activating T-cells undergo necroptotic cell death as an apparent consequence of massive macro-autophagy [124,125]. Normally, macro-autophagy, usually simply referred to as autophagy, is a starvation-induced response wherein a cell ‘eats’ its own organelles to conserve nutrients by enwrapping these organelles in a specialized compartment known as an autophagosome. It also represents a way for a cell to get rid of damaged organelles, such as mitochondria that produce excessive amounts of ROS [126]. Bell et al. have shown that caspase-8 forms a complex with RIPK1 and FADD on the autophagosomal membrane. Formation of this complex appeared to be initiated by the autophagosomal protein Atg5 modified by the ubiquitin-like protein Atg12 [127]. Presumably, the recruitment of RIPK1 leads to its activation in the complex, whereas the role caspase-8 is to inactivate RIPK1 by cleavage. Only in the absence of caspase activation do the cells die as the apparent result of excessive, RIPK1-dependent, autophagy.

4.3 The heterodimer controls necroptosis

If caspase activation is required for development or activation, then how is apoptosis downstream of caspase-8 activity prevented? The answer seems to lie in the caspase-8/FLIPL heterodimer. Some years ago, Dohrman and colleagues demonstrated that FLIPL was essential for proper T-cell development [79]. More recently Leverrier et al. demonstrated that T-cell development in mice deficient in caspase-8 in the T-cell lineage could be rescued by a caspase-8 mutant that could no longer be processed in the inter-linker subunit, but retained catalytic activity [34]. By replacing the DEDs of caspase-8 and FLIPL with inducible dimerization domains, Pop et al. have shown that the most favored way such a caspase-8 species can generate activity, is through heterodimerization with FLIPL [57]. In addition, as mentioned above, this enzyme species has restricted substrate specificity and only poorly induces apoptosis. Several reports have recently been published that demonstrate that the embryonic defect of caspase-8 deficient mice can be rescued by back-crossing these mice on a RIPK3 or RIPK1 deficient background, thus demonstrating the deleterious effects of necroptosis during development [78,128-130]. In one of these reports, Oberst et al. demonstrate that FLIPL expression is essential for preventing necroptosis [78]. This confirms what has been shown previously, that FLIPL is essential for successful T-cell and monocyte differentiation and activation [32,79-85]. At this stage, it is not entirely clear whether the caspase-8/FLIPL heterodimer controls the RIPKs by cleavage or whether cleavage of FLIPL in this complex is essential for signaling towards NF-κB, as has been suggested [79,80,82]. Caspase-8 also influences gene transcription directly by cleaving and inactivation histone deacetylase-7 (HDAC7), a repressor of genes controlled by the MEF2 family of transcription factors [131,132]. Inactivation of HDAC7 in T-cells results in transcriptional upregulation of the orphan nuclear receptor NuR77, which has been associated with their apoptotic demise [133]. However, the MEF2 transcription factors have multiple targets and their activity has been shown to be essential in monocyte differentiation [134], also a process wherein both caspase-8 and FLIPL play a role [81,104]. HDAC7 is the most efficiently cleaved substrate for caspase-8 identified to date, at least in vitro, and is one of very few substrates known to be cleaved by the unprocessed caspase-8/FLIPL heterodimer [57,132]. It may therefore represent an interesting target for further study into the pro-survival role of caspase-8.

4.3 Pro-survival functions of RIPK3?

Although the RIPK3/caspase-8 double knockout mice were born and matured relatively normal, older mice started suffering from a lymphoproliferative disorder, excessive proliferation of the T-cells caused by a lack of T-cell elimination through apoptosis [78,129,130]. This demonstrates the two-faced nature of caspase-8 which, like a game keeper, is on the one hand required to allow successful proliferation of the T-cell population in the first place and on the other hand to cull the herd of T-cells if proliferation gets excessive. Although the observation that the RIPK3 single knockout mice are relatively normal would suggest that RIPK3 is only required for the detrimental death of cells by necroptosis, these mice were also shown have an increased susceptibility to viral infections [99,135], though in a recent report Lu et al. observe no increased susceptibility of RIPK3−/− mice to Murine Herpes Virus [136]. In addition, at least one viral inhibitor of RIPK3 has been identified, indicating that, at least from the viral point of view, activation of RIPK3 is undesirable [135,137]. Finally, the intracellular anti-viral defense has been shown to depend on RIPK1-RIPK3 signaling [99,137,138]. It is as yet unclear whether RIPK3 is only required to execute necroptosis and that this alternate form of cell death itself is what prevents the spread of viral infection or whether the metabolic switch induced by RIPK3 activation is primarily a pro-survival, proliferative or pro-differentiation response, that only turns into necroptotic cell death if a secondary stimulus (e.g. caspase-8 activation) is not received. In a recent report, Biton and Ashkenazi demonstrate that RIPK1, in the absence of caspase activation, has a pro-survival role in the early response upon extensive DNA damage by activating NF-κB through NEMO but induces the production of pro-inflammatory cytokines, such as IL8 and IL6, in a late-phase response through the JNK3 pathway that leads to activation of the transcription factor AP-1 [122]. In addition, they suggest a feed-forward signaling mechanism wherein RIPK1-dependent TNFα production induces cell death in an autocrine manner, as has been suggested previously for L929 cells [139]. In an even more recent paper, the same lab suggests that the same complex also induces TNFα production and subsequent tumor cell death after stimulating the cells with a TNFα related protein, TWEAK [140]. Thus, if RIPK1 is not inactivated by caspase-8 cell death can lead to sterile inflammation, similar to the phenotype observed in mice with caspase-8 deficient keratinocytes [36]. Whether or not RIPK3 has a role in this pathway was not discussed by the authors, since most of the experiments in the Biton study were performed with HeLa cells and these cells do not normally express RIPK3.

5 Post-translational modifications alter caspase-8 activity

5.1 Caspase-8 activity regulated by kinases

Caspase-8 is not only cleaved, but has also been shown to be phosphorylated and ubiquitinated. Several kinases have been shown to phosphorylate caspase-8 and suppress its activation. Whereas caspases-9, -3 and -2 appear to be regulated by serine or threonine phosphorylation, caspase-8 is mostly phosphorylated on a few conserved tyrosine residues. Thus, the serine/threonine kinases RIPK1 and RIPK3 can not control caspase-8 activity. For a detailed overview of the regulation of caspase activity by phosphorylation, we refer the reader to an excellent review by Kurokawa and Kornbluth [141]. It should also be noted here that phosphorylation of Bid, one of the main pro-apoptotic effectors downstream of caspase-8, on a conserved threonine residue in the caspase-8 cleavage motif by one of several kinases [142-146] prevents cleavage and by caspase-8. Considering that the caspase-8/FLIPL heterodimer does not cleave Bid, but that the caspase-10/FLIPL heterodimer does [29,57], phosphorylation of Bid may actually prevent premature induction of apoptosis by caspase-10.

5.2 Caspase-8 activity regulated by ubiquitination

So far we have seen a number of ways in which caspase-8 can remain anti-apoptotic, but how does caspase-8 turn pro-apoptotic? In an insightful study, Jin et al. suggested that caspase-8 undergoes extensive poly-ubiquitination in the DISC. C-terminal ubiquitination of caspase-8 was shown to be mediated by the ubiquitin ligase cullin-3, which was also found to be present in the DISC, downstream of DR4 signaling [147]. Poly-ubiquitination of caspase-8 increased its pro-death potential, apparently by stabilizing the dimers. The deubiquitinating enzyme A20 reversed the process. This is interesting, as A20 has also been shown to inhibit RIPK1 signaling towards NF-κB by removing K63-linked polyubiquitin chains and replacing them with K48-linked chains [120,148,149]. Thus, A20 prevents caspase-8 from becoming pro-apoptotic while at the same time preventing RIPK1 from pro-survival signaling, apparently maintaining the status quo. This C-terminal poly-ubiquitination of caspase-8 observed by Jin et al. may also explain how caspase-8 turns into a pro-apoptotic protein in the first place. After all, as we have seen, unprocessed caspase-8 can only be activated through interaction with FLIPL and, as such, it is incapable of auto-processing [57]. Therefore, a secondary stabilizer, such as a ubiquitin chain, may be required to ‘zip up’ and fully activate the caspase-8 heterodimer.

6 Conclusions

As we have seen, in the initial stages of its activation caspase-8 primarily has non-apoptotic, pro-survival functions. How then, does this good-natured caspase turn into a hardened killer? It all seems to depend on the company it keeps. The heterodimer of caspase-8 and FLIPL is presumed to be formed preferentially upon death receptor ligation; since the Kd for dissociation of the heterodimer is significantly lower than for the homodimer [59,67]. However, the concentration of FLIPL is rate-limiting; upon continued stimulation or when a very strong stimulus is received, homodimers will inevitably form. An alternative heterodimer of caspase-8 with MALT1 has also been suggested to form downstream of antigen receptor stimulation, which appears to have similar properties [150]. Progressive activation of the heterodimer also occurs, as illustrated in Figure 5. If caspase-activity remains absent, either because cells have been treated with a caspase inhibitor, because a viral infection prevents caspase activation or because some other condition prevents the formation of the FADD/caspase-8 DISC, active RIPK1 will eventually execute the cell by necroptosis. FLIPL is good company in this respect, because it actively prevents RIPK1 from executing the cells by a pro-inflammatory type of cell death while simultaneously preventing full activation of caspase-8. In other words, FLIPL keeps the peace and caspase-8 out of trouble. Cleavage of RIPK1 by the caspase-8 partner is essential for this function. Only cleaved FLIPL activates NF-κB, probably by recruiting TRAF2 and cIAP1 to the complex, which leads to K63-linked RIPK1 ubiquitination and interaction of RIPK1 with NEMO, instead of RIPK3 [82,122,151,152]. In this complex, caspase-8 can only be fully activated if it is cleaved by an external protease [57]. If the intrinsic pathway of apoptosis is activated and cytochrome c has been released from the mitochondria, caspase-6, which is activated downstream of caspase-3 and -9, will do so [62]. Alternatively, Cathepsin D appears to cleave caspase-8 upon lysosomal damage [66] while Granzyme B can do the job after a cell has been marked for death by a cytotoxic T-cell [61]. Thus, in a cell that has received extensive damage or has been infected the pro-life/pro-activation signal of the caspase-8/FLIPL heterodimer can still be turned into an apoptotic signal, preventing the spread of an infection or, for example, tumor growth. Activation of the homodimer is similarly controlled from the outside, but this time by poly-ubiquitination of the C-terminus [147]. Altogether, activation of caspase-8 is not primarily a suicide mechanism. This explains the fast, and growing, number of events that rely on caspase-8 activity in the absence of cell death [153-156]. Caspase-8 primarily signals activation and proliferation, only secondary events can turn this signal in a death-inducing mechanism. It is unlikely that RIPK1 and/or RIPK3 are the only relevant substrates of the caspase-8/FLIPL heterodimer. HDAC7 has been mentioned before as an excellent substrate for this heterodimer, but other substrates are likely to turn up [57]. In pathological situations, such as tumor cell proliferation, activation of the heterodimer may be unwanted. Since the active site groove of the heterodimer is not essentially different from the groove in the homodimer, designing specific inhibitors for the heterodimer may be challenging. Active-site directed inhibitors will definitely target both homo- and heterodimers, resulting in necroptosis. Instead, allosteric inhibitors may have to be developed or, alternatively, allosteric activators of caspase-8 that turn the complex from pro-survival into pro-apoptotic, similar to the allosteric activator of caspase-3 recently described by Wolan et al.[157]. Altogether, the insight that caspase-8 can have different functions depending on its dimerization partners has proven to be a crucial insight and significantly improved our understanding of the complex processes of cell death and differentiation. Further research is pending into the exact targets of the heterodimer as compared to the homodimer and, not unimportantly, the role of caspase-10 in the heterodimer.

Figure 5. Progressive processing of caspase-8 determines the outcome of RIPK1-signaling.

Figure 5

1) If caspase-8 is not activated, the complex of RIPK1 (top, orange), FADD (middle, violet) and caspase-8/FLIPL (bottom, green/blue) induces necroptosis, as indicated in the left pane of the figure. 2) Moderate caspase-8 activation in the complex results in FLIPL cleavage, possibly RIPK1 cleavage and signaling towards NF-κB. 3) Cleavage of caspase-8 inter subunit linker in the heterodimer, possibly by an external protease, results in a moderate apoptotic signal and RIPK1 cleavage. 4) Full activation and processing of caspase-8 in the homodimer results in efficient cleavage of RIPK1 and signaling towards apoptosis. Ubiquitination of both RIPK1 and caspase-8 controls the formation of the complex (not in the figure).

Acknowledgements

Bram J. van Raam is supported by a Rubicon fellowship from the Netherlands Organization for Scientific Research (NWO) as well as a fellowship from the Barth Syndrome Foundation. We gratefully acknowledge Christian Zmasek for his insight in the caspase-8 phylogeny and Peter D. Mace for his help in generating the images in Figure 4.

Abbreviations

ALPS

Acute Lympho-Proliferative Syndrome

CARD

Caspase Recruitment Domain

CED

Caenorhabditis elegans Death protein

DD

Death Domain

DED

Death Effector Domain

DISC

Death Inducing Signaling Complex

DR

Death Receptor

FADD

Fas- Associated Death Domain protein

FLIP

FLICE Inhibitory Protein

HDAC

Histone Deacetylase

ICE

Interleukin Converting Enzyme

IL

Interleukin

IRF

Interferon Regulatory Factor

JNK

c-Jun N-terminal Kinase

NEMO

NF-κB Essential Modulator

NF

Nuclear Factor

RIPK

Receptor Interacting Protein Kinase

ROS

Reactive Oxygen Species

TNF

Tumor Necrosis Factor

TNFR

TNF Receptor

TRADD

TNFR Associated Death Domain protein

TRAF

TNFR Associated Factor

TWEAK

TNF-Related Weak inducer of apoptosis

References

  • 1.Thornberry NA, Bull HG, Calaycay JR, Chapman KT, Howard AD, Kostura MJ, Miller DK, Molineaux SM, Weidner JR, Aunins J. A novel heterodimeric cysteine protease is required for interleukin-1 beta processing in monocytes. Nature. 1992;356:768–774. doi: 10.1038/356768a0. [DOI] [PubMed] [Google Scholar]
  • 2.Cerretti DP, Kozlosky CJ, Mosley B, Nelson N, Van NK, Greenstreet TA, March CJ, Kronheim SR, Druck T, Cannizzaro LA. Molecular cloning of the interleukin-1 beta converting enzyme. Science. 1992;256:97–100. doi: 10.1126/science.1373520. [DOI] [PubMed] [Google Scholar]
  • 3.Yuan J, Shaham S, Ledoux S, Ellis HM, Horvitz HR. The C. elegans cell death gene ced-3 encodes a protein similar to mammalian interleukin-1 beta-converting enzyme. Cell. 1993;75:641–652. doi: 10.1016/0092-8674(93)90485-9. [DOI] [PubMed] [Google Scholar]
  • 4.Yuan JY, Horvitz HR. The Caenorhabditis elegans genes ced-3 and ced-4 act cell autonomously to cause programmed cell death. Dev. Biol. 1990;138:33–41. doi: 10.1016/0012-1606(90)90174-h. [DOI] [PubMed] [Google Scholar]
  • 5.Thornberry NA, Molineaux SM. Interleukin-1 beta converting enzyme: a novel cysteine protease required for IL-1 beta production and implicated in programmed cell death. Protein Sci. 1995;4:3–12. doi: 10.1002/pro.5560040102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Pop C, Salvesen GS. Human caspases: Activation, specificity and regulation. J. Biol. Chem. 2009 doi: 10.1074/jbc.R800084200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Fuentes-Prior P, Salvesen GS. The protein structures that shape caspase activity, specificity, activation and inhibition. Biochem. J. 2004;384:201–232. doi: 10.1042/BJ20041142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Kroemer G, Galluzzi L, Vandenabeele P, Abrams J, Alnemri ES, Baehrecke EH, Blagosklonny MV, El-Deiry WS, Golstein P, Green DR, Hengartner M, Knight RA, Kumar S, Lipton SA, Malorni W, Nunez G, Peter ME, Tschopp J, Yuan J, Piacentini M, Zhivotovsky B, Melino G. Classification of cell death: recommendations of the Nomenclature Committee on Cell Death 2009. Cell Death. Differ. 2009;16:3–11. doi: 10.1038/cdd.2008.150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Lindsay J, Esposti MD, Gilmore AP. Bcl-2 proteins and mitochondria-Specificity in membrane targeting for death. Biochim. Biophys. Acta. 2011;1813:532–539. doi: 10.1016/j.bbamcr.2010.10.017. [DOI] [PubMed] [Google Scholar]
  • 10.Kantari C, Walczak H. Caspase-8 and bid: caught in the act between death receptors and mitochondria. Biochim. Biophys. Acta. 2011;1813:558–563. doi: 10.1016/j.bbamcr.2011.01.026. [DOI] [PubMed] [Google Scholar]
  • 11.Degterev A, Hitomi J, Germscheid M, Ch'en IL, Korkina O, Teng X, Abbott D, Cuny GD, Yuan C, Wagner G, Hedrick SM, Gerber SA, Lugovskoy A, Yuan J. Identification of RIP1 kinase as a specific cellular target of necrostatins. Nat. Chem. Biol. 2008;4:313–321. doi: 10.1038/nchembio.83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Declercq W, Vanden Berghe T, Vandenabeele P. RIP kinases at the crossroads of cell death and survival. Cell. 2009;138:229–232. doi: 10.1016/j.cell.2009.07.006. [DOI] [PubMed] [Google Scholar]
  • 13.Lassus P, Opitz-Araya X, Lazebnik Y. Requirement for caspase-2 in stress-induced apoptosis before mitochondrial permeabilization. Science. 2002;297:1352–1354. doi: 10.1126/science.1074721. [DOI] [PubMed] [Google Scholar]
  • 14.Tinel A, Tschopp J. The PIDDosome, a protein complex implicated in activation of caspase-2 in response to genotoxic stress. Science. 2004;304:843–846. doi: 10.1126/science.1095432. [DOI] [PubMed] [Google Scholar]
  • 15.Vakifahmetoglu-Norberg H, Zhivotovsky B. The unpredictable caspase-2: what can it do? Trends Cell Biol. 2010;20:150–159. doi: 10.1016/j.tcb.2009.12.006. [DOI] [PubMed] [Google Scholar]
  • 16.Sohn D, Budach W, Janicke RU. Caspase-2 is required for DNA damage-induced expression of the CDK inhibitor p21(WAF1/CIP1) Cell Death. Differ. 2011 doi: 10.1038/cdd.2011.34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Shi M, Vivian CJ, Lee KJ, Ge C, Morotomi-Yano K, Manzl C, Bock F, Sato S, Tomomori-Sato C, Zhu R, Haug JS, Swanson SK, Washburn MP, Chen DJ, Chen BP, Villunger A, Florens L, Du C. Retraction Notice to: DNA-PKcs-PIDDosome: A Nuclear Caspase-2-Activating Complex with Role in G2/M Checkpoint Maintenance. Cell. 2011;145:161. doi: 10.1016/j.cell.2011.03.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Tiwari M, Lopez-Cruzan M, Morgan WW, Herman B. Loss of caspase-2-dependent apoptosis induces autophagy after mitochondrial oxidative stress in primary cultures of young adult cortical neurons. J. Biol. Chem. 2011;286:8493–8506. doi: 10.1074/jbc.M110.163824. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Shelton SN, Dillard CD, Robertson JD. Activation of caspase-9, but not caspase-2 or caspase-8, is essential for heat-induced apoptosis in Jurkat cells. J. Biol. Chem. 2010;285:40525–40533. doi: 10.1074/jbc.M110.167635. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Denecker G, Ovaere P, Vandenabeele P, Declercq W. Caspase-14 reveals its secrets. J. Cell Biol. 2008;180:451–458. doi: 10.1083/jcb.200709098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Hitomi J, Katayama T, Eguchi Y, Kudo T, Taniguchi M, Koyama Y, Manabe T, Yamagishi S, Bando Y, Imaizumi K, Tsujimoto Y, Tohyama M. Involvement of caspase-4 in endoplasmic reticulum stress-induced apoptosis and Abeta-induced cell death. J Cell Biol. 2004;165:347–356. doi: 10.1083/jcb.200310015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Lakshmanan U, Porter AG. Caspase-4 interacts with TNF receptor-associated factor 6 and mediates lipopolysaccharide-induced NF-kappaB-dependent production of IL-8 and CC chemokine ligand 4 (macrophage-inflammatory protein-1 ) J Immunol. 2007;179:8480–8490. doi: 10.4049/jimmunol.179.12.8480. [DOI] [PubMed] [Google Scholar]
  • 23.Martinon F, Tschopp J. Inflammatory caspases: linking an intracellular innate immune system to autoinflammatory diseases. Cell. 2004;117:561–574. doi: 10.1016/j.cell.2004.05.004. [DOI] [PubMed] [Google Scholar]
  • 24.Miu J, Saleh M, Stevenson MM. Caspase-12 deficiency enhances cytokine responses but does not protect against lethal Plasmodium yoelii 17XL infection. Parasite Immunol. 2010;32:773–778. doi: 10.1111/j.1365-3024.2010.01250.x. [DOI] [PubMed] [Google Scholar]
  • 25.Labbe K, Miu J, Yeretssian G, Serghides L, Tam M, Finney CA, Erdman LK, Goulet ML, Kain KC, Stevenson MM, Saleh M. Caspase-12 dampens the immune response to malaria independently of the inflammasome by targeting NF-kappaB signaling. J. Immunol. 2010;185:5495–5502. doi: 10.4049/jimmunol.1002517. [DOI] [PubMed] [Google Scholar]
  • 26.Wang P, Arjona A, Zhang Y, Sultana H, Dai J, Yang L, LeBlanc PM, Doiron K, Saleh M, Fikrig E. Caspase-12 controls West Nile virus infection via the viral RNA receptor RIG-I. Nat. Immunol. 2010;11:912–919. doi: 10.1038/ni.1933. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Saleh M, Mathison JC, Wolinski MK, Bensinger SJ, Fitzgerald P, Droin N, Ulevitch RJ, Green DR, Nicholson DW. Enhanced bacterial clearance and sepsis resistance in caspase-12-deficient mice. Nature. 2006;440:1064–1068. doi: 10.1038/nature04656. [DOI] [PubMed] [Google Scholar]
  • 28.Los M, Wesselborg S, Schulze-Osthoff K. The role of caspases in development, immunity, and apoptotic signal transduction: lessons from knockout mice. Immunity. 1999;10:629–639. doi: 10.1016/s1074-7613(00)80062-x. [DOI] [PubMed] [Google Scholar]
  • 29.Wachmann K, Pop C, van Raam BJ, Drag M, Mace PD, Snipas SJ, Zmasek C, Schwarzenbacher R, Salvesen GS, Riedl SJ. Activation and specificity of human caspase-10. Biochemistry. 2010;49:8307–8315. doi: 10.1021/bi100968m. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Varfolomeev EE, Schuchmann M, Luria V, Chiannilkulchai N, Beckmann JS, Mett IL, Rebrikov D, Brodianski VM, Kemper OC, Kollet O, Lapidot T, Soffer D, Sobe T, Avraham KB, Goncharov T, Holtmann H, Lonai P, Wallach D. 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. 1998;9:267–276. doi: 10.1016/s1074-7613(00)80609-3. [DOI] [PubMed] [Google Scholar]
  • 31.Yeh WC, Pompa JL, McCurrach ME, Shu HB, Elia AJ, Shahinian A, Ng M, Wakeham A, Khoo W, Mitchell K, El-Deiry WS, Lowe SW, Goeddel DV, Mak TW. FADD: essential for embryo development and signaling from some, but not all, inducers of apoptosis. Science. 1998;279:1954–1958. doi: 10.1126/science.279.5358.1954. [DOI] [PubMed] [Google Scholar]
  • 32.Yeh WC, Itie A, Elia AJ, Ng M, Shu HB, Wakeham A, Mirtsos C, Suzuki N, Bonnard M, Goeddel DV, Mak TW. Requirement for Casper (c-FLIP) in regulation of death receptor-induced apoptosis and embryonic development. Immunity. 2000;12:633–642. doi: 10.1016/s1074-7613(00)80214-9. [DOI] [PubMed] [Google Scholar]
  • 33.Kang TB, Ben-Moshe T, Varfolomeev EE, Pewzner-Jung Y, Yogev N, Jurewicz A, Waisman A, Brenner O, Haffner R, Gustafsson E, Ramakrishnan P, Lapidot T, Wallach D. Caspase-8 serves both apoptotic and nonapoptotic roles. J. Immunol. 2004;173:2976–2984. doi: 10.4049/jimmunol.173.5.2976. [DOI] [PubMed] [Google Scholar]
  • 34.Leverrier S, Salvesen GS, Walsh CM. Enzymatically active single chain caspase-8 maintains T-cell survival during clonal expansion. Cell Death. Differ. 2010 doi: 10.1038/cdd.2010.69. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Mielgo A, Torres VA, Schmid MC, Graf R, Zeitlin SG, Lee P, Shields DJ, Barbero S, Jamora C, Stupack DG. The death effector domains of caspase-8 induce terminal differentiation. PLoS. ONE. 2009;4:e7879. doi: 10.1371/journal.pone.0007879. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Kovalenko A, Kim JC, Kang TB, Rajput A, Bogdanov K, Dittrich-Breiholz O, Kracht M, Brenner O, Wallach D. Caspase-8 deficiency in epidermal keratinocytes triggers an inflammatory skin disease. J. Exp. Med. 2009;206:2161–2177. doi: 10.1084/jem.20090616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Finlay D, Howes A, Vuori K. Critical role for caspase-8 in epidermal growth factor signaling. Cancer Res. 2009;69:5023–5029. doi: 10.1158/0008-5472.CAN-08-3731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Helfer B, Boswell BC, Finlay D, Cipres A, Vuori K, Bong KT, Wallach D, Dorfleutner A, Lahti JM, Flynn DC, Frisch SM. Caspase-8 promotes cell motility and calpain activity under nonapoptotic conditions. Cancer Res. 2006;66:4273–4278. doi: 10.1158/0008-5472.CAN-05-4183. [DOI] [PubMed] [Google Scholar]
  • 39.Chun HJ, Zheng L, Ahmad M, Wang J, Speirs CK, Siegel RM, Dale JK, Puck J, Davis J, Hall CG, Skoda-Smith S, Atkinson TP, Straus SE, Lenardo MJ. Pleiotropic defects in lymphocyte activation caused by caspase-8 mutations lead to human immunodeficiency. Nature. 2002;419:395–399. doi: 10.1038/nature01063. [DOI] [PubMed] [Google Scholar]
  • 40.Kischkel FC, Lawrence DA, Tinel A, LeBlanc H, Virmani A, Schow P, Gazdar A, Blenis J, Arnott D, Ashkenazi A. Death receptor recruitment of endogenous caspase-10 and apoptosis initiation in the absence of caspase-8. J. Biol. Chem. 2001;276:46639–46646. doi: 10.1074/jbc.M105102200. [DOI] [PubMed] [Google Scholar]
  • 41.Sprick MR, Rieser E, Stahl H, Grosse-Wilde A, Weigand MA, Walczak H. Caspase-10 is recruited to and activated at the native TRAIL and CD95 death-inducing signalling complexes in a FADD-dependent manner but can not functionally substitute caspase-8. EMBO J. 2002;21:4520–4530. doi: 10.1093/emboj/cdf441. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Wang J, Chun HJ, Wong W, Spencer DM, Lenardo MJ. Caspase-10 is an initiator caspase in death receptor signaling. Proc. Natl. Acad. Sci. U. S. A. 2001;98:13884–13888. doi: 10.1073/pnas.241358198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Wang J, Zheng L, Lobito A, Chan FK, Dale J, Sneller M, Yao X, Puck JM, Straus SE, Lenardo MJ. Inherited human Caspase 10 mutations underlie defective lymphocyte and dendritic cell apoptosis in autoimmune lymphoproliferative syndrome type II. Cell. 1999;98:47–58. doi: 10.1016/S0092-8674(00)80605-4. [DOI] [PubMed] [Google Scholar]
  • 44.Worth A, Thrasher AJ, Gaspar HB. Autoimmune lymphoproliferative syndrome: molecular basis of disease and clinical phenotype. Br. J. Haematol. 2006;133:124–140. doi: 10.1111/j.1365-2141.2006.05993.x. [DOI] [PubMed] [Google Scholar]
  • 45.Gronbaek K, Dalby T, Zeuthen J, Ralfkiaer E, Guidberg P. The V410I (G1228A) variant of the caspase-10 gene is a common polymorphism of the Danish population. Blood. 2000;95:2184–2185. [PubMed] [Google Scholar]
  • 46.Mace PD, Riedl SJ. Molecular cell death platforms and assemblies. Curr. Opin. Cell Biol. 2010;22:828–836. doi: 10.1016/j.ceb.2010.08.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Pop C, Timmer J, Sperandio S, Salvesen GS. The apoptosome activates caspase-9 by dimerization. Mol. Cell. 2006;22:269–275. doi: 10.1016/j.molcel.2006.03.009. [DOI] [PubMed] [Google Scholar]
  • 48.Boatright KM, Renatus M, Scott FL, Sperandio S, Shin H, Pedersen IM, Ricci JE, Edris WA, Sutherlin DP, Green DR, Salvesen GS. A unified model for apical caspase activation. Mol. Cell. 2003;11:529–541. doi: 10.1016/s1097-2765(03)00051-0. [DOI] [PubMed] [Google Scholar]
  • 49.Scott FL, Stec B, Pop C, Dobaczewska MK, Lee JJ, Monosov E, Robinson H, Salvesen GS, Schwarzenbacher R, Riedl SJ. The Fas-FADD death domain complex structure unravels signalling by receptor clustering. Nature. 2009;457:1019–1022. doi: 10.1038/nature07606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Wang L, Yang JK, Kabaleeswaran V, Rice AJ, Cruz AC, Park AY, Yin Q, Damko E, Jang SB, Raunser S, Robinson CV, Siegel RM, Walz T, Wu H. The Fas-FADD death domain complex structure reveals the basis of DISC assembly and disease mutations. Nat. Struct. Mol. Biol. 2010;17:1324–1329. doi: 10.1038/nsmb.1920. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Tu S, McStay GP, Boucher LM, Mak T, Beere HM, Green DR. In situ trapping of activated initiator caspases reveals a role for caspase-2 in heat shock-induced apoptosis. Nat. Cell Biol. 2006;8:72–77. doi: 10.1038/ncb1340. [DOI] [PubMed] [Google Scholar]
  • 52.Muzio M, Stockwell BR, Stennicke HR, Salvesen GS, Dixit VM. An induced proximity model for caspase-8 activation. J. Biol. Chem. 1998;273:2926–2930. doi: 10.1074/jbc.273.5.2926. [DOI] [PubMed] [Google Scholar]
  • 53.Keller N, Mares J, Zerbe O, Grutter MG. Structural and biochemical studies on procaspase-8: new insights on initiator caspase activation. Structure. 2009;17:438–448. doi: 10.1016/j.str.2008.12.019. [DOI] [PubMed] [Google Scholar]
  • 54.Keller N, Grutter MG, Zerbe O. Studies of the molecular mechanism of caspase-8 activation by solution NMR. Cell Death. Differ. 2010;17:710–718. doi: 10.1038/cdd.2009.155. [DOI] [PubMed] [Google Scholar]
  • 55.Oberst A, Pop C, Tremblay AG, Blais V, Denault JB, Salvesen GS, Green DR. Inducible dimerization and inducible cleavage reveal a requirement for both processes in caspase-8 activation. J. Biol. Chem. 2010;285:16632–16642. doi: 10.1074/jbc.M109.095083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Pop C, Fitzgerald P, Green DR, Salvesen GS. Role of proteolysis in caspase-8 activation and stabilization. Biochemistry. 2007;46:4398–4407. doi: 10.1021/bi602623b. [DOI] [PubMed] [Google Scholar]
  • 57.Pop C, Oberst A, Drag M, van Raam BJ, Riedl SJ, Green DR, Salvesen GS. FLIPL induces caspase 8 activity in the absence of interdomain caspase 8 cleavage and alters substrate specificity. Biochem. J. 2011;433:447–457. doi: 10.1042/BJ20101738. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Chang DW, Xing Z, Capacio VL, Peter ME, Yang X. Interdimer processing mechanism of procaspase-8 activation. EMBO J. 2003;22:4132–4142. doi: 10.1093/emboj/cdg414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Donepudi M, Mac SA, Briand C, Grutter MG. Insights into the regulatory mechanism for caspase-8 activation. Mol. Cell. 2003;11:543–549. doi: 10.1016/s1097-2765(03)00059-5. [DOI] [PubMed] [Google Scholar]
  • 60.Medema JP, Scaffidi C, Kischkel FC, Shevchenko A, Mann M, Krammer PH, Peter ME. FLICE is activated by association with the CD95 death-inducing signaling complex (DISC) EMBO J. 1997;16:2794–2804. doi: 10.1093/emboj/16.10.2794. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Medema JP, Toes RE, Scaffidi C, Zheng TS, Flavell RA, Melief CJ, Peter ME, Offringa R, Krammer PH. Cleavage of FLICE (caspase-8) by granzyme B during cytotoxic T lymphocyte-induced apoptosis. Eur. J. Immunol. 1997;27:3492–3498. doi: 10.1002/eji.1830271250. [DOI] [PubMed] [Google Scholar]
  • 62.Cowling V, Downward J. Caspase-6 is the direct activator of caspase-8 in the cytochrome c-induced apoptosis pathway: absolute requirement for removal of caspase-6 prodomain. Cell Death. Differ. 2002;9:1046–1056. doi: 10.1038/sj.cdd.4401065. [DOI] [PubMed] [Google Scholar]
  • 63.Schug ZT, Gonzalvez F, Houtkooper RH, Vaz FM, Gottlieb E. BID is cleaved by caspase-8 within a native complex on the mitochondrial membrane. Cell Death. Differ. 2010 doi: 10.1038/cdd.2010.135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Gonzalvez F, Schug ZT, Houtkooper RH, MacKenzie ED, Brooks DG, Wanders RJ, Petit PX, Vaz FM, Gottlieb E. Cardiolipin provides an essential activating platform for caspase-8 on mitochondria. J. Cell Biol. 2008;183:681–696. doi: 10.1083/jcb.200803129. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Lutter M, Fang M, Luo X, Nishijima M, Xie X, Wang X. Cardiolipin provides specificity for targeting of tBid to mitochondria. Nat. Cell Biol. 2000;2:754–761. doi: 10.1038/35036395. [DOI] [PubMed] [Google Scholar]
  • 66.Conus S, Perozzo R, Reinheckel T, Peters C, Scapozza L, Yousefi S, Simon HU. Caspase-8 is activated by cathepsin D initiating neutrophil apoptosis during the resolution of inflammation. J Exp. Med. 2008;205:685–698. doi: 10.1084/jem.20072152. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Boatright KM, Deis C, Denault JB, Sutherlin DP, Salvesen GS. Activation of caspases-8 and -10 by FLIP(L) Biochem. J. 2004;382:651–657. doi: 10.1042/BJ20040809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Dohrman A, Russell JQ, Cuenin S, Fortner K, Tschopp J, Budd RC. Cellular FLIP long form augments caspase activity and death of T cells through heterodimerization with and activation of caspase-8. J. Immunol. 2005;175:311–318. doi: 10.4049/jimmunol.175.1.311. [DOI] [PubMed] [Google Scholar]
  • 69.Micheau O, Thome M, Schneider P, Holler N, Tschopp J, Nicholson DW, Briand C, Grutter MG. The long form of FLIP is an activator of caspase-8 at the Fas death-inducing signaling complex. J. Biol. Chem. 2002;277:45162–45171. doi: 10.1074/jbc.M206882200. [DOI] [PubMed] [Google Scholar]
  • 70.Yu JW, Jeffrey PD, Shi Y. Mechanism of procaspase-8 activation by c-FLIPL. Proc. Natl. Acad. Sci. U. S. A. 2009;106:8169–8174. doi: 10.1073/pnas.0812453106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Hughes MA, Harper N, Butterworth M, Cain K, Cohen GM, MacFarlane M. Reconstitution of the death-inducing signaling complex reveals a substrate switch that determines CD95-mediated death or survival. Mol. Cell. 2009;35:265–279. doi: 10.1016/j.molcel.2009.06.012. [DOI] [PubMed] [Google Scholar]
  • 72.Han DKM, Chaudhary PM, Wright ME, Friedman C, Trask BJ, Riedel RT, Baskin DG, Schwartz SM, Hood L. MRIT, a novel death-effector domain-containing protein, interacts with caspases and Bcl-XL and initiates cell death. Proc Natl Acad Sci USA. 1997;94 doi: 10.1073/pnas.94.21.11333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Hu S, Vincenz C, Ni J, Gentz R, Dixit VM. I-FLICE, a novel inhibitor of tumor necrosis factor receptor-1- and CD-95-induced apoptosis. J Biol Chem. 1997;272 doi: 10.1074/jbc.272.28.17255. [DOI] [PubMed] [Google Scholar]
  • 74.Rasper DM, Vaillancourt JP, Hadano S, Houtzager VM, Seiden I, Keen SL, Tawa P, Xanthoudakis S, Nasir J, Martindale D, Koop BF, Peterson EP, Thornberry NA, Huang J, MacPherson DP, Black SC, Hornung F, Lenardo MJ, Hayden MR, Roy S, Nicholson DW. Cell death attenuation by 'Usurpin', a mammalian DED-caspase homologue that precludes caspase-8 recruitment and activation by the CD-95 (Fas, APO-1) receptor complex. Cell Death Differ. 1998;5 doi: 10.1038/sj.cdd.4400370. [DOI] [PubMed] [Google Scholar]
  • 75.Shu HB, Halpin DR, Goeddel DV. Casper is a FADD- and caspase-related inducer of apoptosis. Immunity. 1997;6 doi: 10.1016/s1074-7613(00)80450-1. [DOI] [PubMed] [Google Scholar]
  • 76.Tschopp J, Irmler M, Thome M. Inhibition of fas death signals by FLIPs. Curr. Opin. Immunol. 1998;10:552–558. doi: 10.1016/s0952-7915(98)80223-9. [DOI] [PubMed] [Google Scholar]
  • 77.Scaffidi C, Schmitz I, Krammer PH, Peter ME. The role of c-FLIP in modulation of CD95-induced apoptosis. J. Biol. Chem. 1999;274:1541–1548. doi: 10.1074/jbc.274.3.1541. [DOI] [PubMed] [Google Scholar]
  • 78.Oberst A, Dillon CP, Weinlich R, McCormick LL, Fitzgerald P, Pop C, Hakem R, Salvesen GS, Green DR. Catalytic activity of the caspase-8-FLIP(L) complex inhibits RIPK3-dependent necrosis. Nature. 2011;471:363–367. doi: 10.1038/nature09852. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Dohrman A, Kataoka T, Cuenin S, Russell JQ, Tschopp J, Budd RC. Cellular FLIP (long form) regulates CD8+ T cell activation through caspase-8-dependent NF-kappa B activation. J. Immunol. 2005;174:5270–5278. doi: 10.4049/jimmunol.174.9.5270. [DOI] [PubMed] [Google Scholar]
  • 80.Golks A, Brenner D, Krammer PH, Lavrik IN. The c-FLIP-NH2 terminus (p22-FLIP) induces NF-kappaB activation. J. Exp. Med. 2006;203:1295–1305. doi: 10.1084/jem.20051556. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Huang QQ, Perlman H, Huang Z, Birkett R, Kan L, Agrawal H, Misharin A, Gurbuxani S, Crispino JD, Pope RM. FLIP: a novel regulator of macrophage differentiation and granulocyte homeostasis. Blood. 2010;116:4968–4977. doi: 10.1182/blood-2009-11-252841. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Kataoka T, Tschopp J. N-terminal fragment of c-FLIP(L) processed by caspase 8 specifically interacts with TRAF2 and induces activation of the NF-kappaB signaling pathway. Mol. Cell Biol. 2004;24:2627–2636. doi: 10.1128/MCB.24.7.2627-2636.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Kataoka T, Budd RC, Holler N, Thome M, Martinon F, Irmler M, Burns K, Hahne M, Kennedy N, Kovacsovics M, Tschopp J. The caspase-8 inhibitor FLIP promotes activation of NF-kappaB and Erk signaling pathways. Curr. Biol. 2000;10:640–648. doi: 10.1016/s0960-9822(00)00512-1. [DOI] [PubMed] [Google Scholar]
  • 84.Lens SM, Kataoka T, Fortner KA, Tinel A, Ferrero I, MacDonald RH, Hahne M, Beermann F, Attinger A, Orbea HA, Budd RC, Tschopp J. The caspase 8 inhibitor c-FLIP(L) modulates T-cell receptor-induced proliferation but not activation-induced cell death of lymphocytes. Mol. Cell Biol. 2002;22:5419–5433. doi: 10.1128/MCB.22.15.5419-5433.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Thome M, Tschopp J. Regulation of lymphocyte proliferation and death by FLIP. Nat. Rev. Immunol. 2001;1:50–58. doi: 10.1038/35095508. [DOI] [PubMed] [Google Scholar]
  • 86.Wu W, Rinaldi L, Fortner KA, Russell JQ, Tschopp J, Irvin C, Budd RC. Cellular FLIP long form-transgenic mice manifest a Th2 cytokine bias and enhanced allergic airway inflammation. J. Immunol. 2004;172:4724–4732. doi: 10.4049/jimmunol.172.8.4724. [DOI] [PubMed] [Google Scholar]
  • 87.Blanchard H, Donepudi M, Tschopp M, Kodandapani L, Wu JC, Grutter MG. Caspase-8 specificity probed at subsite S(4): crystal structure of the caspase-8-Z-DEVD-cho complex. J. Mol. Biol. 2000;302:9–16. doi: 10.1006/jmbi.2000.4041. [DOI] [PubMed] [Google Scholar]
  • 88.Blanchard H, Kodandapani L, Mittl PR, Marco SD, Krebs JF, Wu JC, Tomaselli KJ, Grutter MG. The three-dimensional structure of caspase-8: an initiator enzyme in apoptosis. Structure. 1999;7:1125–1133. doi: 10.1016/s0969-2126(99)80179-8. [DOI] [PubMed] [Google Scholar]
  • 89.Watt W, Koeplinger KA, Mildner AM, Heinrikson RL, Tomasselli AG, Watenpaugh KD. The atomic-resolution structure of human caspase-8, a key activator of apoptosis. Structure. 1999;7:1135–1143. doi: 10.1016/s0969-2126(99)80180-4. [DOI] [PubMed] [Google Scholar]
  • 90.Xu G, Cirilli M, Huang Y, Rich RL, Myszka DG, Wu H. Covalent inhibition revealed by the crystal structure of the caspase-8/p35 complex. Nature. 2001;410:494–497. doi: 10.1038/35068604. [DOI] [PubMed] [Google Scholar]
  • 91.Ekici OD, Li ZZ, Campbell AJ, James KE, Asgian JL, Mikolajczyk J, Salvesen GS, Ganesan R, Jelakovic S, Grutter MG, Powers JC. Design, synthesis, and evaluation of aza-peptide Michael acceptors as selective and potent inhibitors of caspases-2, -3, -6, -7, -8, -9, and -10. J. Med. Chem. 2006;49:5728–5749. doi: 10.1021/jm0601405. [DOI] [PubMed] [Google Scholar]
  • 92.Vaux DL. Caspases and apoptosis - biology and terminology. Cell Death. Differ. 1999;6:493–494. doi: 10.1038/sj.cdd.4400523. [DOI] [PubMed] [Google Scholar]
  • 93.Vercammen D, Beyaert R, Denecker G, Goossens V, van LG, Declercq W, Grooten J, Fiers W, Vandenabeele P. Inhibition of caspases increases the sensitivity of L929 cells to necrosis mediated by tumor necrosis factor. J. Exp. Med. 1998;187:1477–1485. doi: 10.1084/jem.187.9.1477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Schulze-Osthoff K, Bakker AC, Vanhaesebroeck B, Beyaert R, Jacob WA, Fiers W. Cytotoxic activity of tumor necrosis factor is mediated by early damage of mitochondrial functions. Evidence for the involvement of mitochondrial radical generation. J. Biol. Chem. 1992;267:5317–5323. [PubMed] [Google Scholar]
  • 95.Denecker G, Vercammen D, Declercq W, Vandenabeele P. Apoptotic and necrotic cell death induced by death domain receptors. Cell Mol. Life Sci. 2001;58:356–370. doi: 10.1007/PL00000863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Zhou Q, Snipas S, Orth K, Muzio M, Dixit VM, Salvesen GS. Target protease specificity of the viral serpin CrmA. Analysis of five caspases. J. Biol. Chem. 1997;272:7797–7800. doi: 10.1074/jbc.272.12.7797. [DOI] [PubMed] [Google Scholar]
  • 97.Degterev A, Huang Z, Boyce M, Li Y, Jagtap P, Mizushima N, Cuny GD, Mitchison TJ, Moskowitz MA, Yuan J. Chemical inhibitor of nonapoptotic cell death with therapeutic potential for ischemic brain injury. Nat. Chem. Biol. 2005;1:112–119. doi: 10.1038/nchembio711. [DOI] [PubMed] [Google Scholar]
  • 98.Walsh CM, Wen BG, Chinnaiyan AM, O'Rourke K, Dixit VM, Hedrick SM. A role for FADD in T cell activation and development. Immunity. 1998;8:439–449. doi: 10.1016/s1074-7613(00)80549-x. [DOI] [PubMed] [Google Scholar]
  • 99.Cho YS, Challa S, Moquin D, Genga R, Ray TD, Guildford M, Chan FK. Phosphorylation-driven assembly of the RIP1-RIP3 complex regulates programmed necrosis and virus-induced inflammation. Cell. 2009;137:1112–1123. doi: 10.1016/j.cell.2009.05.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.He S, Wang L, Miao L, Wang T, Du F, Zhao L, Wang X. Receptor interacting protein kinase-3 determines cellular necrotic response to TNF-alpha. Cell. 2009;137:1100–1111. doi: 10.1016/j.cell.2009.05.021. [DOI] [PubMed] [Google Scholar]
  • 101.Zhang DW, Shao J, Lin J, Zhang N, Lu BJ, Lin SC, Dong MQ, Han J. RIP3, an energy metabolism regulator that switches TNF-induced cell death from apoptosis to necrosis. Science. 2009;325:332–336. doi: 10.1126/science.1172308. [DOI] [PubMed] [Google Scholar]
  • 102.Vanlangenakker N, Vanden Berghe T, Bogaert P, Laukens B, Zobel K, Deshayes K, Vucic D, Fulda S, Vandenabeele P, Bertrand MJ. cIAP1 and TAK1 protect cells from TNF-induced necrosis by preventing RIP1/RIP3-dependent reactive oxygen species production. Cell Death. Differ. 2011;18:656–665. doi: 10.1038/cdd.2010.138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Feng S, Yang Y, Mei Y, Ma L, Zhu DE, Hoti N, Castanares M, Wu M. Cleavage of RIP3 inactivates its caspase-independent apoptosis pathway by removal of kinase domain. Cell Signal. 2007;19:2056–2067. doi: 10.1016/j.cellsig.2007.05.016. [DOI] [PubMed] [Google Scholar]
  • 104.Rebe C, Cathelin S, Launay S, Filomenko R, Prevotat L, L'Ollivier C, Gyan E, Micheau O, Grant S, Dubart-Kupperschmitt A, Fontenay M, Solary E. Caspase-8 prevents sustained activation of NF-kappaB in monocytes undergoing macrophagic differentiation. Blood. 2007;109:1442–1450. doi: 10.1182/blood-2006-03-011585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Lin Y, Devin A, Cook A, Keane MM, Kelliher M, Lipkowitz S, Liu ZG. The death domain kinase RIP is essential for TRAIL (Apo2L)-induced activation of IkappaB kinase and c-Jun N-terminal kinase. Mol. Cell Biol. 2000;20:6638–6645. doi: 10.1128/mcb.20.18.6638-6645.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Martinon F, Holler N, Richard C, Tschopp J. Activation of a pro-apoptotic amplification loop through inhibition of NF-kappaB-dependent survival signals by caspase-mediated inactivation of RIP. FEBS Lett. 2000;468:134–136. doi: 10.1016/s0014-5793(00)01212-6. [DOI] [PubMed] [Google Scholar]
  • 107.Stanger BZ, Leder P, Lee TH, Kim E, Seed B. RIP: a novel protein containing a death domain that interacts with Fas/APO-1 (CD95) in yeast and causes cell death. Cell. 1995;81:513–523. doi: 10.1016/0092-8674(95)90072-1. [DOI] [PubMed] [Google Scholar]
  • 108.Wong WW, Gentle IE, Nachbur U, Anderton H, Vaux DL, Silke J. RIPK1 is not essential for TNFR1-induced activation of NF-kappaB. Cell Death. Differ. 2010;17:482–487. doi: 10.1038/cdd.2009.178. [DOI] [PubMed] [Google Scholar]
  • 109.Ting AT, Pimentel-Muinos FX, Seed B. RIP mediates tumor necrosis factor receptor 1 activation of NF-kappaB but not Fas/APO-1-initiated apoptosis. EMBO J. 1996;15:6189–6196. [PMC free article] [PubMed] [Google Scholar]
  • 110.Micheau O, Tschopp J. Induction of TNF receptor I-mediated apoptosis via two sequential signaling complexes. Cell. 2003;114:181–190. doi: 10.1016/s0092-8674(03)00521-x. [DOI] [PubMed] [Google Scholar]
  • 111.Werner MH, Wu C, Walsh CM. Emerging roles for the death adaptor FADD in death receptor avidity and cell cycle regulation. Cell Cycle. 2006;5:2332–2338. doi: 10.4161/cc.5.20.3385. [DOI] [PubMed] [Google Scholar]
  • 112.Walsh CM, Edinger AL. The complex interplay between autophagy, apoptosis, and necrotic signals promotes T-cell homeostasis. Immunol. Rev. 2010;236:95–109. doi: 10.1111/j.1600-065X.2010.00919.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Zheng L, Bidere N, Staudt D, Cubre A, Orenstein J, Chan FK, Lenardo M. Competitive control of independent programs of tumor necrosis factor receptor-induced cell death by TRADD and RIP1. Mol. Cell Biol. 2006;26:3505–3513. doi: 10.1128/MCB.26.9.3505-3513.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Enesa K, Zakkar M, Chaudhury H, Luong IA, Rawlinson L, Mason JC, Haskard DO, Dean JL, Evans PC. NF-kappaB suppression by the deubiquitinating enzyme Cezanne: a novel negative feedback loop in pro-inflammatory signaling. J. Biol. Chem. 2008;283:7036–7045. doi: 10.1074/jbc.M708690200. [DOI] [PubMed] [Google Scholar]
  • 115.Komander D, Barford D. Structure of the A20 OTU domain and mechanistic insights into deubiquitination. Biochem. J. 2008;409:77–85. doi: 10.1042/BJ20071399. [DOI] [PubMed] [Google Scholar]
  • 116.Bertrand MJ, Milutinovic S, Dickson KM, Ho WC, Boudreault A, Durkin J, Gillard JW, Jaquith JB, Morris SJ, Barker PA. cIAP1 and cIAP2 facilitate cancer cell survival by functioning as E3 ligases that promote RIP1 ubiquitination. Mol. Cell. 2008;30:689–700. doi: 10.1016/j.molcel.2008.05.014. [DOI] [PubMed] [Google Scholar]
  • 117.Dynek JN, Goncharov T, Dueber EC, Fedorova AV, Izrael-Tomasevic A, Phu L, Helgason E, Fairbrother WJ, Deshayes K, Kirkpatrick DS, Vucic D. c-IAP1 and UbcH5 promote K11-linked polyubiquitination of RIP1 in TNF signalling. EMBO J. 2010;29:4198–4209. doi: 10.1038/emboj.2010.300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Gerlach B, Cordier SM, Schmukle AC, Emmerich CH, Rieser E, Haas TL, Webb AI, Rickard JA, Anderton H, Wong WW, Nachbur U, Gangoda L, Warnken U, Purcell AW, Silke J, Walczak H. Linear ubiquitination prevents inflammation and regulates immune signalling. Nature. 2011;471:591–596. doi: 10.1038/nature09816. [DOI] [PubMed] [Google Scholar]
  • 119.Newton K, Matsumoto ML, Wertz IE, Kirkpatrick DS, Lill JR, Tan J, Dugger D, Gordon N, Sidhu SS, Fellouse FA, Komuves L, French DM, Ferrando RE, Lam C, Compaan D, Yu C, Bosanac I, Hymowitz SG, Kelley RF, Dixit VM. Ubiquitin chain editing revealed by polyubiquitin linkage-specific antibodies. Cell. 2008;134:668–678. doi: 10.1016/j.cell.2008.07.039. [DOI] [PubMed] [Google Scholar]
  • 120.Wertz IE, O'Rourke KM, Zhou H, Eby M, Aravind L, Seshagiri S, Wu P, Wiesmann C, Baker R, Boone DL, Ma A, Koonin EV, Dixit VM. De-ubiquitination and ubiquitin ligase domains of A20 downregulate NF-kappaB signalling. Nature. 2004;430:694–699. doi: 10.1038/nature02794. [DOI] [PubMed] [Google Scholar]
  • 121.Vandenabeele P, Declercq W, Van HF, Vanden Berghe T. The role of the kinases RIP1 and RIP3 in TNF-induced necrosis. Sci. Signal. 2010;3:re4. doi: 10.1126/scisignal.3115re4. [DOI] [PubMed] [Google Scholar]
  • 122.Biton S, Ashkenazi A. NEMO and RIP1 Control Cell Fate in Response to Extensive DNA Damage via TNF-alpha Feedforward Signaling. Cell. 2011;145:92–103. doi: 10.1016/j.cell.2011.02.023. [DOI] [PubMed] [Google Scholar]
  • 123.Osborn SL, Diehl G, Han SJ, Xue L, Kurd N, Hsieh K, Cado D, Robey EA, Winoto A. Fas-associated death domain (FADD) is a negative regulator of T-cell receptor-mediated necroptosis. Proc. Natl. Acad. Sci. U. S. A. 2010;107:13034–13039. doi: 10.1073/pnas.1005997107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Bell BD, Walsh CM. Coordinate regulation of autophagy and apoptosis in T cells by death effectors: FADD or foundation. Autophagy. 2009;5:238–240. doi: 10.4161/auto.5.2.7512. [DOI] [PubMed] [Google Scholar]
  • 125.Ch'en IL, Beisner DR, Degterev A, Lynch C, Yuan J, Hoffmann A, Hedrick SM. Antigen-mediated T cell expansion regulated by parallel pathways of death. Proc. Natl. Acad. Sci. U. S. A. 2008;105:17463–17468. doi: 10.1073/pnas.0808043105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Gottlieb RA, Carreira RS. Autophagy in health and disease. 5. Mitophagy as a way of life. Am. J. Physiol Cell Physiol. 2010;299:C203–C210. doi: 10.1152/ajpcell.00097.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Bell BD, Leverrier S, Weist BM, Newton RH, Arechiga AF, Luhrs KA, Morrissette NS, Walsh CM. FADD and caspase-8 control the outcome of autophagic signaling in proliferating T cells. Proc. Natl. Acad. Sci. U. S. A. 2008;105:16677–16682. doi: 10.1073/pnas.0808597105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Zhang H, Zhou X, McQuade T, Li J, Chan FK, Zhang J. Functional complementation between FADD and RIP1 in embryos and lymphocytes. Nature. 2011;471:373–376. doi: 10.1038/nature09878. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Kaiser WJ, Upton JW, Long AB, Livingston-Rosanoff D, Daley-Bauer LP, Hakem R, Caspary T, Mocarski ES. RIP3 mediates the embryonic lethality of caspase-8-deficient mice. Nature. 2011;471:368–372. doi: 10.1038/nature09857. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Ch'en IL, Tsau JS, Molkentin JD, Komatsu M, Hedrick SM. Mechanisms of necroptosis in T cells. J. Exp. Med. 2011;208:633–641. doi: 10.1084/jem.20110251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Verdin E, Dequiedt F, Kasler HG. Class II histone deacetylases: versatile regulators. Trends Genet. 2003;19:286–293. doi: 10.1016/S0168-9525(03)00073-8. [DOI] [PubMed] [Google Scholar]
  • 132.Scott FL, Fuchs GJ, Boyd SE, Denault JB, Hawkins CJ, Dequiedt F, Salvesen GS. Caspase-8 cleaves histone deacetylase 7 and abolishes its transcription repressor function. J. Biol. Chem. 2008;283:19499–19510. doi: 10.1074/jbc.M800331200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.He YW. Orphan nuclear receptors in T lymphocyte development. J. Leukoc. Biol. 2002;72:440–446. [PubMed] [Google Scholar]
  • 134.Aude-Garcia C, Collin-Faure V, Bausinger H, Hanau D, Rabilloud T, Lemercier C. Dual roles for MEF2A and MEF2D during human macrophage terminal differentiation and c-Jun expression. Biochem. J. 2010;430:237–244. doi: 10.1042/BJ20100131. [DOI] [PubMed] [Google Scholar]
  • 135.Upton JW, Kaiser WJ, Mocarski ES. Virus inhibition of RIP3-dependent necrosis. Cell Host. Microbe. 2010;7:302–313. doi: 10.1016/j.chom.2010.03.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Lu JV, Weist BM, van Raam BJ, Marro BS, Srinivas P, Bell BD, Luhrs KA, Lane TE, Salvesen GS, Walsh CM. Complementary roles of FADD and RIPK3 in T cell homeostasis and antiviral immunity. Proc Natl Acad Sci USA Accepted for publication. 2010 doi: 10.1073/pnas.1102779108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Rebsamen M, Heinz LX, Meylan E, Michallet MC, Schroder K, Hofmann K, Vazquez J, Benedict CA, Tschopp J. DAI/ZBP1 recruits RIP1 and RIP3 through RIP homotypic interaction motifs to activate NF-kappaB. EMBO Rep. 2009 doi: 10.1038/embor.2009.109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Michallet MC, Meylan E, Ermolaeva MA, Vazquez J, Rebsamen M, Curran J, Poeck H, Bscheider M, Hartmann G, Konig M, Kalinke U, Pasparakis M, Tschopp J. TRADD protein is an essential component of the RIG-like helicase antiviral pathway. Immunity. 2008;28:651–661. doi: 10.1016/j.immuni.2008.03.013. [DOI] [PubMed] [Google Scholar]
  • 139.Wu YT, Tan HL, Huang Q, Sun XJ, Zhu X, Shen HM. zVAD-induced necroptosis in L929 cells depends on autocrine production of TNFalpha mediated by the PKC-MAPKs-AP-1 pathway. Cell Death. Differ. 2011;18:26–37. doi: 10.1038/cdd.2010.72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Ikner A, Ashkenazi A. TWEAK induces apoptosis through a death-signaling complex comprising receptor-interacting protein 1 (RIP1), Fas-associated death domain (FADD) and caspase-8. J. Biol. Chem. 2011 doi: 10.1074/jbc.M110.203745. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Kurokawa M, Kornbluth S. Caspases and kinases in a death grip. Cell. 2009;138:838–854. doi: 10.1016/j.cell.2009.08.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Hellwig CT, Ludwig-Galezowska AH, Concannon CG, Litchfield DW, Prehn JH, Rehm M. Activity of protein kinase CK2 uncouples Bid cleavage from caspase-8 activation. J. Cell Sci. 2010;123:1401–1406. doi: 10.1242/jcs.061143. [DOI] [PubMed] [Google Scholar]
  • 143.Desagher S, Osen-Sand A, Montessuit S, Magnenat E, Vilbois F, Hochmann A, Journot L, Antonsson B, Martinou JC. Phosphorylation of bid by casein kinases I and II regulates its cleavage by caspase 8. Mol. Cell. 2001;8:601–611. doi: 10.1016/s1097-2765(01)00335-5. [DOI] [PubMed] [Google Scholar]
  • 144.Holmstrom TH, Schmitz I, Soderstrom TS, Poukkula M, Johnson VL, Chow SC, Krammer PH, Eriksson JE. MAPK/ERK signaling in activated T cells inhibits CD95/Fas-mediated apoptosis downstream of DISC assembly. EMBO J. 2000;19:5418–5428. doi: 10.1093/emboj/19.20.5418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Kamer I, Sarig R, Zaltsman Y, Niv H, Oberkovitz G, Regev L, Haimovich G, Lerenthal Y, Marcellus RC, Gross A. Proapoptotic BID is an ATM effector in the DNA-damage response. Cell. 2005;122:593–603. doi: 10.1016/j.cell.2005.06.014. [DOI] [PubMed] [Google Scholar]
  • 146.Vogel A, Aslan JE, Willenbring H, Klein C, Finegold M, Mount H, Thomas G, Grompe M. Sustained phosphorylation of Bid is a marker for resistance to Fas-induced apoptosis during chronic liver diseases. Gastroenterology. 2006;130:104–119. doi: 10.1053/j.gastro.2005.10.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Jin Z, Li Y, Pitti R, Lawrence D, Pham VC, Lill JR, Ashkenazi A. Cullin3-Based Polyubiquitination and p62-Dependent Aggregation of Caspase-8 Mediate Extrinsic Apoptosis Signaling. Cell. 2009 doi: 10.1016/j.cell.2009.03.015. [DOI] [PubMed] [Google Scholar]
  • 148.He KL, Ting AT. A20 inhibits tumor necrosis factor (TNF) alpha-induced apoptosis by disrupting recruitment of TRADD and RIP to the TNF receptor 1 complex in Jurkat T cells. Mol. Cell Biol. 2002;22:6034–6045. doi: 10.1128/MCB.22.17.6034-6045.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Shembade N, Ma A, Harhaj EW. Inhibition of NF-kappaB signaling by A20 through disruption of ubiquitin enzyme complexes. Science. 2010;327:1135–1139. doi: 10.1126/science.1182364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Kawadler H, Gantz MA, Riley JL, Yang X. The paracaspase MALT1 controls caspase-8 activation during lymphocyte proliferation. Mol. Cell. 2008;31:415–421. doi: 10.1016/j.molcel.2008.06.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Vince JE, Pantaki D, Feltham R, Mace PD, Cordier SM, Schmukle AC, Davidson AJ, Callus BA, Wong WW, Gentle IE, Carter H, Lee EF, Walczak H, Day CL, Vaux DL, Silke J. TRAF2 must bind to cellular inhibitors of apoptosis for tumor necrosis factor (tnf) to efficiently activate nf-{kappa}b and to prevent tnf-induced apoptosis. J. Biol. Chem. 2009;284:35906–35915. doi: 10.1074/jbc.M109.072256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Mace PD, Linke K, Feltham R, Schumacher FR, Smith CA, Vaux DL, Silke J, Day CL. Structures of the cIAP2 RING domain reveal conformational changes associated with ubiquitin-conjugating enzyme (E2) recruitment. J. Biol. Chem. 2008;283:31633–31640. doi: 10.1074/jbc.M804753200. [DOI] [PubMed] [Google Scholar]
  • 153.Knox PG, Davies CC, Ioannou M, Eliopoulos AG. The death domain kinase RIP1 links the immunoregulatory CD40 receptor to apoptotic signaling in carcinomas. J. Cell Biol. 2011;192:391–399. doi: 10.1083/jcb.201003087. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Chen L, Park SM, Tumanov AV, Hau A, Sawada K, Feig C, Turner JR, Fu YX, Romero IL, Lengyel E, Peter ME. CD95 promotes tumour growth. Nature. 2010;465:492–496. doi: 10.1038/nature09075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Lemmers B, Salmena L, Bidere N, Su H, Matysiak-Zablocki E, Murakami K, Ohashi PS, Jurisicova A, Lenardo M, Hakem R, Hakem A. Essential role for caspase-8 in Toll-like receptors and NFkappaB signaling. J. Biol. Chem. 2007;282:7416–7423. doi: 10.1074/jbc.M606721200. [DOI] [PubMed] [Google Scholar]
  • 156.Su H, Bidere N, Zheng L, Cubre A, Sakai K, Dale J, Salmena L, Hakem R, Straus S, Lenardo M. Requirement for caspase-8 in NF-kappaB activation by antigen receptor. Science. 2005;307:1465–1468. doi: 10.1126/science.1104765. [DOI] [PubMed] [Google Scholar]
  • 157.Wolan DW, Zorn JA, Gray DC, Wells JA. Small-molecule activators of a proenzyme. Science. 2009;326:853–858. doi: 10.1126/science.1177585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Eckhart L, Ballaun C, Hermann M, VandeBerg JL, Sipos W, Uthman A, Fischer H, Tschachler E. Identification of novel mammalian caspases reveals an important role of gene loss in shaping the human caspase repertoire. Mol. Biol. Evol. 2008;25:831–841. doi: 10.1093/molbev/msn012. [DOI] [PubMed] [Google Scholar]

RESOURCES