Skip to main content
Cellular and Molecular Life Sciences: CMLS logoLink to Cellular and Molecular Life Sciences: CMLS
. 2010 Feb 19;67(10):1607–1618. doi: 10.1007/s00018-010-0287-9

Non-caspase proteases: triggers or amplifiers of apoptosis?

Karen Schrader 1,2, Jisen Huai 1, Lars Jöckel 1,2,3, Carolin Oberle 1,5, Christoph Borner 1,2,3,4,
PMCID: PMC11115756  PMID: 20169397

Abstract

Caspases are the most important effectors of apoptosis, the major form of programmed cell death (PCD) in multicellular organisms. This is best reflected by the appearance of serious development defects in mice deficient for caspase-8, -9, and -3. Meanwhile, caspase-independent PCD, mediated by other proteases or signaling components has been described in numerous publications. Although we do not doubt that such cell death exists, we propose that it has evolved later during evolution and is most likely not designed to execute, but to amplify and speed-up caspase-dependent cell death. This review shall provide evidence for such a concept.

Keywords: Caspases, Serine proteases, Cathepsins, Apoptosis, Necrosis, Programmed cell death, Caspase-independent

Introduction

Programmed cell death (PCD) is a controlled process to eliminate used-up, damaged, or misplaced cells during the embryonic development and the tissue homeostasis of multicellular organisms [1]. Aberrant PCD can lead to embryonic defects or diseases such as neurodegeneration, immunodeficiency, cancer, or autoimmunity. Apoptosis, the most abundant form of PCD, is a morphologically defined process characterized by cell shrinkage, plasma membrane blebbing, nuclear condensation and fragmentation, the formation of membrane enclosed apoptotic bodies and their selective uptake by phagocytes without provoking inflammatory responses [2]. Necrosis is another form of cell death that has also been characterized as programmed under certain circumstances (called necroptosis), but which ultimately leads to cell lysis and the triggering of an inflammatory response [3]. For a detailed comparison of molecular mechanisms that control apoptosis and necrosis/necroptosis, read Van Herreweghe et al. in this issue. In this review, we will focus on apoptotic and necrotic PCD based on the morphologies defined by Kerr and Wyllie [2].

It has become clear over the years that cysteine aspartyl proteases (caspases) are the major drivers of apoptotic PCD [4]. Some morphological features such as the fragmentation of genomic DNA into nucleosome-sized fragments strictly depend on caspases because the major endonuclease CAD is activated by caspase-mediated destruction of its inhibitor [5]. However, other aspects of apoptosis, such as cell shrinkage, membrane blebbing and the surface exposure of phosphatidylserine, one of several possible ligands or “eat-me” signals for phagocytosis, can still occur in the apparent absence of caspase activation [6]. These aspects are clearly distinct from necrosis and may therefore be classified as caspase-independent, apoptosis-like PCD. The problem is that caspase-independence has been largely defined by the cellular treatment with so-called broad-range caspase inhibitors such as ZVAD.fmk and QVD-OPh [7]. In other words, it has been assumed that these inhibitors completely inhibit all 10–12 caspases in mammals in a way that even low residual caspase activities would not be able to trigger apoptosis. Unfortunately, with the best of our knowledge, we cannot make sure that this is the case. First, although the inhibitory constants (K i) of Z-VAD.fmk have been determined for all recombinant caspases in vitro, they vary between the different caspases, and it is unclear if caspase inhibition is as effective inside cells as it is in vitro [8, 9]. Second, Z-VAD.fmk also inhibits other proteases, such as calpains and cathepsins, especially at the typical doses used for caspase inhibition (100 μM) [9, 10]. Third, general caspase inhibitors may get rapidly degraded inside the cells. Finally, the threshold of active effector caspase molecules still capable of executing apoptosis has not yet been determined. To definitely exclude minor caspase activities inside cells, we would have to genetically knock-out, in an inducible manner, all ten caspases in mice. Although this is a heroic approach, it is the only way to prove that caspase-independent, apoptosis-like PCD distinct from necrosis indeed exists.

Despite these uncertainties, it has become evident that the cell death observed under caspase inhibiting conditions is rather slow and inefficient [6]. This may translate into a delayed fashion to phagocytose apoptotic bodies in vivo, which of course can influence cell homeostasis and contribute to diseases. Thus, whatever mechanism is involved, PCD under low caspase-activating or caspase-inhibiting conditions may be a crucial physiological or pathological process in multicellular organisms in vivo. This is why it is important to understand the underlying mechanisms, for example if non-caspase proteases are implicated in human diseases and may hence become targets of intervention in addition to caspases.

The importance of caspases for PCD in vivo and in vitro

Lower eukaryotes are ideal model systems to study the importance of caspases in PCD. In the nematode C. elegans the deletion of the sole executioner caspase CED-3 results in the survival of all 131 cells, which would otherwise die during embryonic development [11]. CED-3 is also required for physiological and DNA damage-induced cell death in the worm germline [12]. Caspase-like proteins and caspase-independent mediators have been identified in worms [13], but these enzymes may amplify rather than execute CED-3 mediated death signaling as we propose for higher eukaroytes (see below). Nevertheless, C. elegans also exhibits a caspase-independent, necrotic program to eliminate neuronal cells by hyperactive ion channels [14]. As far as we can extract from the literature, caspase-independent PCD has been rarely described in Drosophila melanogaster. Here, cells perfectly survive apoptosis induction in the absence of caspase activity [15], although recent studies have demonstrated alternative cell death pathways which may be mediated via other proteases [16]. But again this might be due to residual caspase activities which are amplified by the action of other proteases. The failure of cells, in both C. elegans and D. melanogaster, to undergo caspase-independent PCD is probably due to the absence of mitochondrial outer membrane permeabilization (MOMP), a feature that has been adopted by higher eukaryotes later in evolution [7].

In contrast to worms and flies, mice and men express 10 and 12 caspases, respectively [4, 17]. They are classified into initiator caspases, acting early in the apoptotic pathway and effector caspases, performing the execution function. Initiator caspases contain large prodomains, called CARD or DED, which serve the function to bring two monomeric inactive caspases in close proximity for autoactivation when an adapter binds to the prodomain [18]. Although this close proximity already activates the initiator caspase, it can further process itself to become more active, change its localization or inactivate itself, depending on the context [19]. Effector caspases do not have large prodomains, already exist as dimers, and require proteolytic cleavage by an initiator caspase to remove an inhibitory loop covering the active site [20]. Effector caspases then can autoprocess to an active tetramer consisting of two small and two large subunits. While effector caspases can probably accept hundreds of cellular substrates whose cleavages lead to the apoptotic morphology [21], initiator caspases have a very limited substrate spectrum, predominantly effector caspases.

Two major caspase-dependent signaling pathways are known in mammals. In the extrinsic death receptor pathway, extracellular TNF-like ligands such as TNFα, FasL or TRAIL (also see Weinlich et al. in this issue) bind to cognate receptors on the cell surface, triggering receptor multimerization and a conformational change on the cytoplasmic face of the receptor [22, 23]. This allows the recruitment of the adaptor FADD which assembles the initiator pro-caspase-8 on the receptor. Proximity activated caspase-8 cleaves and activates the effector caspases-3 and -7 [18, 22]. The second signaling pathway, called the mitochondrial or intrinsic pathway (also see Pradelli et al. in this issue), is stimulated by many apoptotic stimuli such a growth factor/cytokine removal, DNA-damaging drugs and irradiation, viruses and the detachment from the extracellular matrix (anoikis). By still unknown mechanisms these stimuli are sensed by a subgroup of Bcl-2 family members, the BH3-only proteins, which, through transcriptional upregulation, phosphorylation or proteolytic cleavage stimulate the oligomerization of Bax and Bak, another class of pro-apoptotic Bcl-2 proteins on mitochondria [2426]. This somehow increases the permeability of the outer mitochondrial membrane (MOMP) leading to the release of apoptogenic factors from the intermembrane space. Cytochrome c is the major caspase-driving protein of these factors because once in the cytosol it binds the adaptor Apaf-1 which, through ATP hydrolysis and conformational change, recruits monomeric pro-caspase-9 to a platform, called the apoptosome [27, 28]. The apoptosome then processes effector caspases-3 and -7. Importantly, Bax/Bak induced MOMP is required for the intrinsic pathway, as Bax/Bak double knock-out (DKO) mice are embryonic lethal, and cells from these mice are highly resistant to various apoptotic stimuli [29, 30]. This is why Bax and Bak have been called the “gateway”, “commitment point”, or “life-or-death-decision point” of apoptosis [30].

All these findings indicate that caspases are indispensable for apoptosis induction via both the extrinsic or the intrinsic pathways. Is this really the case?

Caspase-8 and -10

Several studies demonstrated the essential role of caspase-8 in death receptor-mediated apoptosis. Caspase-8−/− mice are prenatally lethal, show impaired heart muscle development, congested accumulation of erythrocytes and circulatory failure [31, 32]. In addition, human individuals with homozygous caspase-8 reduction-of-function mutations manifest autoimmune lymphoproliferative syndrome (ALPS) [33, 34], a disease found in patients with defective Fas signaling [35].

So, is there apoptosis or cell death in the absence of caspase-8? In mouse cells caspase-dependent apoptosis in response to FasL or TNFα is blocked when caspase-8 is missing [23]. However, FasL and TNFα can also trigger necroptosis [3, 36], a necrotic cell death attributed to massive upregulation of ROS [37] and most likely mediated by receptor-interacting protein-1 or -3 [36, 38, 39]. Unfortunately, to unravel this alternative death pathway, Z-VAD.fmk was used again, and it is unclear if all intracellular caspases were blocked by this inhibitor. Moreover, humans, but not mice, express caspase-10 which can partially overtake caspase-8 function and still mediate caspase-dependent death receptor signaling in human cells. This is probably why loss-of-function mutations of caspase-8 are not as devastating in humans as in mice.

Caspase-9 and -3/-7

Caspase-9-deficient mice die perinatally due to defective brain development associated with decreased apoptosis, and MEFs derived from these animals are resistant to apoptosis induced by stimuli which use the intrinsic, mitochondrial signaling pathway [40, 41]. Most caspase-3−/− mice die peri- or postnatally in mixed 129/SvJ and C57BL/6 background but are viable in C57BL/6 background where they exhibit decreased apoptosis in the brain leading to hyperplasia [42]. Caspase-7−/− are viable and show a mild apoptosis defect, for example in endotoxin-induced lymphocyte apoptosis [43]. However, caspase-3/7 DKO mice die perinatally and MEFs derived from these mice are resistant to apoptosis [43].

Based on the knock-out phenotypes, the caspase-9/Apaf-1/caspase-3/-7 signaling pathway plays a dominant role in intrinsic apoptosis. However, some caspase-deficient cells clearly survive to birth and although cells from these knock-out strains (MEFS) show impaired apoptosis, the death is only delayed, but not abolished [4043]. This indicates that cells can die through an intrinsic pathway independent of the apoptosome and/or caspase-3/-7 activation. Is this pathway truly caspase-independent and does it lead to necrosis or an apoptosis-like phenotype?

To answer this question one has to have a closer look at the Bax/Bak-mediated perforation of the mitochondrial outer membrane (MOMP), the “commitment point” of this pathway. It is intriguing that the phenotype of mice lacking Bax and Bak is much more severe than that unable to activate caspases downstream of MOMP (caspase-9−/− and Apaf-1−/−) [29, 30]. This indicates that Bax/Bak do not only trigger the cytochrome c-mediated caspase-9/Apaf-1 signaling branch but also other signaling pathways which are activated due to MOMP. These pathways are likely to be responsible for the cell death which is still observed in apoptosome- and/or caspase-3/-7-deficient cells and animals. Besides cytochrome c, numerous other intermembrane space proteins are released after MOMP, such as Smac/Diablo [44, 45], HtrA2/Omi [46, 47], endonuclease G [48] and apoptosis-inducing factor (AIF) [49]. Various groups have proposed that these proteins can actively induce caspase-independent PCD following mitochondrial release [50]. However, apart from Smac/Diablo and HtrA2/Omi, which facilitate caspase-dependent apoptosis by sequestering XIAP, the other apoptogenic factors have not been convincingly shown to execute caspase-independent cell death. Instead, they are needed for vital functions inside mitochondria and their lack triggers a sort of ROS-driven necrotic response.

In summary, since AIF, endonuclease G or HtrA2/Omi are not clearly involved in executing cell death in the presence of Z-VAD.fmk or in caspase-9- or caspase-3/-7-deficient cells, can other caspases take over the apoptotic job under these circumstances?

Caspases-1, -4/5/11, -12, and -14

Caspases-1, -4, -5, and -11 are enzymes mainly involved in inflammatory responses. Caspase-1 is present in both humans and mice, while caspase-4 and -5 are human enzymes replaced by caspase-11 in the mouse [51]. Caspase-5 and caspase-1 are components of the NALP1 inflammasome, a complex involved in caspase-1 activation [52]. Caspase-1 activation processes the pro-inflammatory cytokines IL-1β and IL-18 [5254]. Both caspase-11−/− and -1−/− mice develop normally and do not show major defects in apoptosis regulation [53, 54]. A form of cell death, called pyroptosis has been described to be associated with caspase-1 activation [55]. However, apart from this, none of the inflammatory caspases seem to mimic or regulate caspases-9, -3 and -7 in the intrinsic pathway. The same is true for caspase-12 and -14. Caspase-12 negatively regulates caspase-1 activation [56], and caspase-14 is crucial for the terminal differentiation of human keratinocytes and cornification [57].

Caspases-2 and -6

Caspase-2 is thought to be the closest homolog of nematodal CED-3 [58]. Despite many reports regarding the function of caspase-2 in a variety of apoptotic processes, the precise role of caspase-2 in apoptosis remains to be verified in vivo, because caspase-2−/− mice display few, if any apoptotic defects [59, 60]. Structurally, caspase-2 is classified as initiator caspase and its activation platform seems to be the PIDDosome, a protein complex containing PIDD (p53-inducible protein with a death domain), an adaptor protein RAIDD (RIP-associated ICH-1) and caspase-2 [6163]. Overexpression of PIDD results in spontaneous activation of caspase-2 and sensitization of the cells to genotoxic stress-induced apoptosis [61]. Substrate profiling studies revealed that caspase-2 cannot process any other member of the caspase family, but can cleave the Bcl-2 family member Bid, presumably to stimulate cytochrome c release [64]. In this respect, caspase-2 could replace the initiating activity of caspase-8 on the type II mitochondrial pathway, but this has not yet been tested, for example by generating caspase-2/-8 DKO mice. Caspase-2/-9 DKO mice exhibit a striking similarity to those lacking caspase-9 [65]. Moreover, caspase-9−/− cells fail to process caspase-2, and pro-caspase-2 activation was shown to be dependent on caspase-3 [65, 66]. All these findings place caspase-2 activation downstream of the apoptosome and caspase-3 activation and suggest that it is an amplifier rather than an initiator caspase.

Caspase-6 poses another puzzle in the caspase field. Due to its short pro-domain and similar substrate specificity as caspase-3 and -7, it is considered an effector caspase. However, caspase-6−/− mice are essentially normal and cells from these mice have not revealed any apoptotic defect so far [67]. But maybe caspase-6 could replace caspase-3/-7 when they are missing, a possibility that has not yet been addressed experimentally. Rather it was shown that caspase-6 is processed and activated by caspase-3 leading to an amplification loop of the caspase cascade [66, 68] In the final stage of this cascade caspase-6 catalyzes the activation of caspase-8 and -10 [69].

Non-caspase proteases as cell death regulators

Although we do not entirely know if cells deficient of particular caspases or treated with Z-VAD.fmk or QVD-OPh die by necrosis or use caspase backup systems or low residual caspase activities to die, it is clear that this cell death still displays proteolytic degradation features. This suggests the role of alternative proteolytic machineries in cell death. Many proteases, including calpains, serine proteases, metalloproteases, cathepsins and the proteasome have been found to be activated in response to apoptotic stimuli. Here, we will focus on two enzyme families, the serine proteases [70, 71] and the cathepsins [72, 73] to illustrate that they primarily act as amplifiers of caspase cascades rather than as initiators or effectors of caspase-independent cell death.

Serine proteases: triggers of MOMP and effector caspase-3 processing

Serine proteases were first implicated in apoptosis in 1987, when it was shown that serine protease inhibitors delayed apoptosis of melanoma cells [74]. By 1994, it was demonstrated that the introduction of chymotrypsin or trypsin into tumor cells led to cell death reminiscent of apoptosis [75]. Later Abate and Schroder [76] found that LPS caused a concentration-dependent toxicity in a macrophage cell line, which was inhibited by the serine protease inhibitors TPCK and TLCK. These investigators did not attempt to isolate or identify the apoptotic serine proteases. In parallel, Masson and Tschopp [77] discovered in 1987 that a family of serine proteases, the granzymes, were capable of inducing cell lysis together with perforin. The expression of granzymes was however restricted to cytotoxic T (CTL), natural killer (NK) and mast cells, and it took another 5 years until it was recognized that these serine proteases induced classical features of apoptosis [78, 79]. In 2003, a novel trypsin-like serine protease, called EOS was isolated from macrophages [80], and we and others suggested the implication of one or several serine proteases in ER-stress and DNA damage induced apoptosis in a variety of eukaryotic cells [6, 81].

Granzymes and cell death

Granzymes (Gzm) are serine proteases that play a crucial role in eliminating virally infected and malignant cells in cooperation with the FasL-Fas signaling system [79, 82, 83]. They are present in the cytotoxic granules of CTLs and NK cells. Upon contact with target cells, these enzymes are released into the intermembrane space together with the pore-forming protein perforin and are delivered to the target cells by a still not entirely understood mechanism [79, 83]. Gzm A and B are the most abundant and extensively studied members of the granzyme family, and studies in CTLs from Gzm A−/−, Gzm B−/− or Gzm A/B DKO cells confirmed that they independently and synergistically induce cell death through distinct apoptotic pathways [84].

Gzm B cleaves substrates following aspartate residues, thus exhibiting a substrate specificity similar to that of caspase family members. It can engage multiple components of the apoptotic machinery in target cells [83, 84]. By directly processing and activating caspase-3 and -7 it majorly produces ROS, the exposure of PS and a decrease in the mitochondrial membrane potential [84, 85]. Via cleavage of the BH3-only protein Bid and the Bcl-2 survival factor Mcl-1, it triggers Bax/Bak mediated cytochrome c release and caspase-3 activation indirectly as well as a fall in the mitochondrial membrane potential [84, 8688]. But even when both pathways are blocked, i.e., in Bax/Bak DKO cells in the presence of Z-VAD.fmk or QVD-OPh, the target cells still die in a so-called MOMP- and caspase-independent manner due to Gzm B action [84]. This might be due to the direct cleavage of apoptosis-relevant substrates by Gzm B [79].

Gzm A is a highly selective tryptic protease that was initially found to trigger a rapid form of cell death that exhibits all of the morphological features associated with apoptosis: membrane blebbing, chromatin condensation and nuclear fragmentation [79]. In contrast to Gzm B, Gzm A does not activate caspases and neither Bcl-2 overexpression nor Z-VAD.fmk treatment prevented GzmA induced cell death [89]. It was proposed that Gzm A may act via ROS production at mitochondrial respiration complexes [84, 89] and the induction of DNA nicks in the nucleus via the so-called SET complex [90]. However, very recently, Metkar et al. [91] convincingly showed that the killing effect of Gzm A is probably due to artificially high concentrations of the protease (micromolar) used in most in vitro experiments. Lower concentrations of human Gzm A (nanomolar) stimulate monocytic cells to secrete pro-inflammatory cytokines (IL-1β, TNFα and IL-6) and thereby provokes a pro-inflammatory response in vivo to resist LPS-induced toxicity. Thus, the Gzm A/B pair may have evolved to amplify FasL-mediated apoptosis of transformed and virally infected cells by combining Gzm B-mediated caspase activation with Gzm A-mediated ROS production and a pro-inflammatory response.

Apoptotic serine proteases in non-CTL/NK cells and their amplifying role at early and late stages of apoptosis

The essential role of granzymes in effective CTL- and NK-mediated cell killing suggests that other cell types may also exploit a serine protease-based mechanism to amplify caspase-mediated apoptosis. A few number of candidate serine proteases have so far been identified, HtrA2/Omi, AP24, a serine protease activated by Fas signaling of ca. 95 kD, three chymotrypsin-like proteases, called p16, p50 and p60 and serine proteases implicated in ER stress and DNA-damage induced apoptosis.

HtrA2/Omi, a mitochondrial survival protease

As discussed above HtrA2/Omi is a survival rather than an apoptotic serine protease. While in the intermembrane space, this protease protects cells from Bax-mediated cytochrome c release and apoptosis [92, 93].

AP24 and Fas-activated protease, two proteases triggering DNA fragmentation at a later step

DNA fragmentation was long thought to be a crucial, early event of apoptosis, occurring in the dying cell before it is recognized and engulfed by macrophages. However, recent data indicate that apoptotic endonucleases (CAD, DNase II) mainly act at later stages of the death process, when the apoptotic bodies are already removed by phagocytosis [94]. In addition to caspase-activated endonuclease CAD, serine proteases may be important in activating other endonucleases in order to accelerate this removal/degradation process [95, 96]. Indeed, a cytosolic serine protease activity was isolated from FasL-treated Jurkat T cells and shown to induce DNA fragmentation [97]. Moreover, during PBOX-6-induced cell death of CML cells where oligonucleosomal DNA fragmentation takes place in the absence of caspase and CAD activation, a chymotrypsin-like serine protease was found to be required for the activation of a manganese-dependent acidic endonuclease [98]. Finally, also during necrotic cell death, serine proteases have been implicated in DNA fragmentation [96]. Unfortunately, none of these serine proteases have yet been cloned.

AP24 is a 24-kD protease with an elastase-like activity that triggers oligonucleosomal DNA fragmentation indirectly by inactivating a serpin, called LEI, through translational modification and converting it into an endonuclease (l-DNase II) [99]. The protease is activated in response to a variety of stimuli, including TNFα, UV irradiation and chemotherapeutic drugs [100, 101], and it is regulated by sphingomyelin [102], calcium/calmodulin-dependent kinase II [103] and members of the Bcl-2 family [104]. However, the exact mode of action of the protease, in particular, how it induces apoptosis, affects MOMP and amplifies caspase-3 activation has remained obscure.

Chymotrypsin-like protease, called p16, p50 and p60 and a ER-stress activated serine protease: amplifiers of apoptosis at different stages

In addition to being involved in later stages of apoptosis, such as DNA fragmentation, serine proteases have been found to act at various stages of caspase-mediated apoptotic signaling. For example, (1) apoptosome-stimulated caspase-9 processing is preventable by serine protease inhibitors [105], (2) caspase-3 processing and activation can at least partially be prevented by the inhibition of chymotrypsin-like proteases [6, 81, 106] and (3) during ER-stress one or several serine proteases are activated which act both upstream of mitochondria to stimulate cytochrome c release as well as downstream of mitochondria to further process and activate caspase-3 [6]. Since the cleavage fragments of caspase-3 generated by serine proteases are similar to those generated by caspase-8 or -9 (p20, p17), the serine protease(s) in question may have aspartyl activities like Gzm B.

p16 is a 16-kD chymotrypsin-like protease that is specifically activated by staurosporine in AML cells [106]. It seems to contribute to the apoptotic morphology, regulation of caspase-3, altered nuclear morphology and DNA degradation, although the amino acid sequence and exact site of action have not yet been determined. p50 and p60 were isolated from Jurkat T cells and are activated in response to staurosporine [107]. Interestingly, they have been shown to act at a late stage of apoptosis, downstream of caspase-3, probably by accelerating nuclear fragmentation and condensation as proposed above for p24 and the Fas-activated serine protease. Consistent with this notion, both serine proteases seem to be activated in the cytosol and then translocate to the nucleus as apoptosis progresses.

In 2003, we reported on the activation of one or several serine protease in response to the ER stressing drugs BFA, tunicamycin and thapsigargin in three different cellular systems, fibroblasts, HeLa cells and FDC-P1 monocytes [6]. The serine proteases exhibited three sites of action. First, they were important for the release of cytochrome c from mitochondria as this release was blocked with the broad range serine protease inhibitor AEBSF/Pefabloc. On the other hand they were responsible, at least in part, for caspase-3 processing as caspase-3 was still processed to the p20/p17 fragments in the presence of Z-VAD.fmk although no caspase-3 activity could be measured. Third, serine proteases seemed to contribute to phagocytosis as PS exposure and the uptake of apoptotic bodies by macrophages in vitro was still evident in Z-VAD.fmk-treated, but blocked in AEBSF-treated cells. Strikingly, while AEBSF could delay apoptosis, it could not inhibit it completely, and clonogenic survival was not possible after removing the apoptotic stimulus. This indicates that ER-stress induced apoptosis is still majorly driven by caspases, but serine proteases amplify this process by acting up- and downstream of mitochondria. We still await the isolation of the serine protease(s) in order to identify the substrate target between ER stress and MOMP.

Serpins, the endogenous inhibitors of serine proteases: can they tell us anything about apoptosis regulation by serine proteases?

Studies demonstrating the effect of the naturally occurring serine protease inhibitors (serpins) in regulating apoptosis also provide indirect evidence for the role of serine proteases in this process [108]. The viral serpin CrmA was originally discovered through its ability to prevent IL-1 processing by inhibiting caspase-1 [109]. Since then CrmA was shown to inhibit activated caspase-8 as well as Gzm B and hence prevent both Fas/TNF- and Gzm B-mediated apoptosis [110112]. A more potent inhibition of Gzm B is achieved by the serpin PI-9 which is the only human protein with such a specificity [113]. Other members of the serpin family such as PAI-2 (plasminogen activator inhibitor 2), PN-1 (protease nexin 1) and Spi2A have also been implicated in the regulation of cell death [70, 71], and as mentioned above, AP24 induces DNA fragmentation by inactivating the serpin LEI, converting its elastase inhibiting activity to a l-DNaseII activity.

Lysosomal cathepsins: are they early triggers of PCD?

Numerous reports have implicated lysosomal cathepsins in early stages of caspase-dependent and -independent PCD [72, 73]. But are these proteases really initiators of cell death? This is a crucial issue as it has been recently proposed that cathepsins could be new therapeutic targets in immune responses by regulating the life span of inflammatory, T and B cells [72].

Cathepsins encompass three classes of lysosomal proteases, the serine proteases cathepsins A and G, the aspartic proteases cathepsins D and E and the 11, so far known human cysteine proteases cathepsins B, C, F, H, K, L, O, S, V, W and X/Z [72, 73]. Most of the cathepsins are endopeptidases, with the exception of cathepsins C and X/Z. These enzymes are all synthesized as inactive proenzymes, posttranslationally modified by glycosylations and targeted to the lumen of the lysosomes via the mannose-6-phosphate receptor sorting pathway. The processing of cathepsins from the inactive to their catalytically active form mainly occurs in the lysosome [114]. Therefore, upon lysosomal membrane permeability (LMP), which seems to widely occur in apoptotic cells, cathepsins are released in their processed form.

Cathepsins can be active in the cytosol at neutral pH, but the life-time of these enzymes is limited due to unfolding-induced inactivation [115]. In addition, most cells express in the cytosol potent cathepsin inhibitors, so called stefins or cystatins [115, 116]. So how would cathepsins then be able to trigger cellular processes in the cytosol, such as apoptosis? First, cathepsins B, D and L, the enzymes mostly implicated in apoptosis are more stable than the other isoforms. Second, stefins and cystatins may be inactivated during apoptosis. Third, the local pH around damaged lysosomes may be acidic enough to transiently stimulate cathepsin-mediated substrate cleavage and the propagation of an apoptotic signal through the cytoplasm [117].

A prerequisite for the proapoptotic function of cathepsins is that they must be released from the lysosome into the cytoplasm by lysosomal membrane permeabilization (LMP) [118]. The major problem is that the vast majority of LMP experiments have been performed with so called lysosomotropic agents such as l-leucyl-l-leucine methyl ester (Leu-Leu-OMe) which directly perforate the lysosomal membrane [118, 119]. Any effect that is observed after this treatment is potentially non-physiological. What such experiments can tell us is how cathepsins could impart on cell survival once they appear in the cytosol.

The best characterized cytosolic substrate for cathepsins to date is the pro-apoptotic Bcl-2 family member Bid [120]. Cleavage of Bid by cathepsins B, L, K and S leads to the formation of tBid which can translocate to mitochondria to trigger Bax/Bak-induced MOMP [29, 30, 121, 122]. However, Bid is unlikely to be the only cellular substrate of cathepsins in this pathway. When mice lacking the major intracellular inhibitor of cysteine cathepsins, stefin B, were crossed with Bid−/− mice, no rescue of spontaneous cathepins-dependent neuronal apoptosis was observed [123]. Similarly, Bid−/− MEFs were not protected against MOMP induced by lysosomotropic photosensitizers [118]. Indeed, very recently, the group of Boris Turk identified pro-apoptotic Bak and BimEL and anti-apoptotic Bcl-2, Bcl-xL and Mcl-1 as additional cysteine cathepsin substrates [124]. However, since Leu-Leu-OMe was used again in these studies, it is unclear if these cleavages also occur in response to apoptotic stimuli. Cathepsins can also directly process and activate caspases such as pro-caspase-3 and -7 [120, 125, 126]. Moreover, in vitro cleavage of pro-caspase-2 by cathepsin B generated a fragment showing cytochrome c releasing activity [127]. Also, during spontaneous apoptosis of neutrophils cathepsin D is released from azurophilic granules, leading to a direct and death receptor-independent activation of caspase-8 [128]. Therefore, direction activation of caspases by cathepsins may have physiological roles in some cells, such as neutrophils.

Whereas the findings described above point towards a rapid, early induction of LMP by apoptotic stimuli, several other reports propose that lysosomes are not the primary target of these stimuli. For example, in response to TNFα, FasL or TRAIL, LMP was found to be downstream of caspase-8 activation suggesting that caspases are responsible for the release of cathepsins B and D into the cytosol [129133]. Specifically, caspase-8 appeared to activate caspase-9, and caspase-9, but no Apaf-1 was required for LMP induced by TNFα [134]. Unfortunately, in most of these experiments, cause and effect between LMP and caspase activation and/or MOMP have not been accurately dissected due to the lack of appropriate knock-out systems and the use of rather non-specific caspase-8 and -2 inhibitors.

If LMP is a decisive, early event in the signaling cascade of apoptosis, cathepsin release and action must also be critical for physiology and pathology in vivo. A variety of cathepsin deficient mouse strains have been generated. Depending on the cathepsin deletion, these mice exhibit aberrancies in bone remodeling, keratinocyte differentiation, carcinogenesis, antigen processing and immune regulation in general [135]. These findings indicate that cathepsins are not just boring enzymes which degrade bulk proteins in lysosomes but may have specific cellular functions. However, characteristic apoptosis defects are rarely seen in cathepsin knock-out animals, with the exception of a lack of resolution of inflammation due to neutrophil accumulation in cathepsin D knock-outs [128], reduced keratinocyte apoptosis resulting in periodic hair loss in cathepsin L deficient mice [136, 137] and diminished liver damage upon TNFα challenge in mice deficient of cathepsin B [130]. Of course this may be due to redundancies in the system, so that only double or triple cathepsin KO mice would reveal gross abnormalities in vivo. But we rather speculate that LMP and the subsequent release of cathepsins into the cytosol are late events that amplify rather than trigger apoptosis. In some cases such as the resolution from inflammation, this amplification may be crucial to rapidly kill neutrophils [128], but in other cases, it may not be relevant because MOMP and caspase activation can already do the job effectively.

Why has no one attempted to dissect the order of this signaling pathway, for example by investigating if LMP and cathepsin release still occur in cells deficient for Bax/Bak, components of the apoptosome (Apaf-1 or caspase-9) or the major effectors caspase-3 and -7? The only data pointing towards a function of LMP and cathepsin release downstream of Bax/Bak was published by the group of Gregory Gores, where they found that Bax can translocate to a fraction enriched in lysosomes during staurosporine induced apoptosis [138]. However, it was not shown if it was really the lysosomally associated, and not the mitochondrial Bax, which mediated this effect. Moreover, it is evident from the literature that Bax−/− cells are not particularly resistant to staurosporine-induced cell death [29, 30]. By contrast, cells deficient in apoptosomal components are resistant to apoptosis induced by staurosporine and other stimuli [40, 41], and hence LMP could be the result of apoptosome formation after Bax/Bak-mediated MOMP. By using two different cell systems (monocytes and fibroblasts) deficient in Bax and/or Bak, caspases or Apaf-1, exposed to four different apoptotic stimuli (FasL, etoposide, UV and IL-3 removal), we could recently show that LMP and cathepsin release required Bax/Bak and the apoptosome and therefore occurred downstream of MOMP [139].

Conclusions

There is no doubt that serine proteases, cathepsins and other non-caspase proteases are emerging as key players of PCD. However, we think that instead of initiating/triggering an apoptotic response, they rather support or propagate caspase-mediated apoptosis signaling upstream, downstream or aside of mitochondria, provoke late events such as oligonucleosomal or nicked DNA fragmentation or phagocytosis or regulate caspase-independent death processes such as necrosis. What is now required is a more accurate, unbiased analysis of where exactly these proteases act in the apoptotic signaling pathway and what steps they indeed control. In addition, for serine proteases it will be important to develop methods and approaches that will permit the precise identification of serine proteases involved in cell death (apoptosis and necrosis). Only then will we be capable of judging if novel therapeutic drugs directed against these proteases are worth being developed in order to cure degenerative diseases.

Acknowledgments

Our work is supported by the Spemann Graduate School of Biology and Medicine (SGBM, GSC-4) and the Centre for Biological Signaling Studies (bioss, EXC 294), both supported by the Excellence Initiative of the German Federal and State Governments, and the Deutsche Forschungsgemeinschaft (BO-1933 and GRK1104).

References

  • 1.Jacobson MD, Weil M, Raff MC. Programmed cell death in animal development. Cell. 1997;88:347–354. doi: 10.1016/S0092-8674(00)81873-5. [DOI] [PubMed] [Google Scholar]
  • 2.Kerr JF, Wyllie AH, Currie AR. Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics. Br J Cancer. 1972;26:239–257. doi: 10.1038/bjc.1972.33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.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]
  • 4.Li J, Yuan J. Caspases in apoptosis and beyond. Oncogene. 2008;27:6194–6206. doi: 10.1038/onc.2008.297. [DOI] [PubMed] [Google Scholar]
  • 5.Enari M, Sakahira H, Yokoyama H, Okawa K, Iwamatsu A, Nagata S. A caspase-activated DNase that degrades DNA during apoptosis, and its inhibitor ICAD. Nature. 1998;391:43–50. doi: 10.1038/34112. [DOI] [PubMed] [Google Scholar]
  • 6.Egger L, Schneider J, Rheme C, Tapernoux M, Hacki J, Borner C. Serine proteases mediate apoptosis-like cell death and phagocytosis under caspase-inhibiting conditions. Cell Death Differ. 2003;10:1188–1203. doi: 10.1038/sj.cdd.4401288. [DOI] [PubMed] [Google Scholar]
  • 7.Tait SWG, Green DR. Caspase-independent cell death: leaving the set without the final cut. Oncogene. 2008;27:6452–6461. doi: 10.1038/onc.2008.311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Garcia-Calvo M, Peterson EP, Leiting B, Ruel R, Nicholson DW, Thornberry NA. Inhibition of human caspases by peptide-based and macromolecular inhibitors. J Biol Chem. 1998;273:32608–32613. doi: 10.1074/jbc.273.49.32608. [DOI] [PubMed] [Google Scholar]
  • 9.Chauvier D, Ankri D, Charriaut-Marlangue C, Casimir R, Jacotot E. Broad-spectrum caspase inhibitors: from myth to reality? Cell Death Differ. 2007;14:387–391. doi: 10.1038/sj.cdd.4402044. [DOI] [PubMed] [Google Scholar]
  • 10.Schotte P, Declercq W, Van Huffel S, Vandenabeele P, Beyaert R. Non-specific effects of methylketone peptide inhibitors of caspases. FEBS Lett. 1998;442:117–121. doi: 10.1016/S0014-5793(98)01640-8. [DOI] [PubMed] [Google Scholar]
  • 11.Ellis HM, Horvitz HR. Genetic control of programmed cell death in the nematode C. elegans . Cell. 1986;44:817–829. doi: 10.1016/0092-8674(86)90004-8. [DOI] [PubMed] [Google Scholar]
  • 12.Gartner A, Milstein S, Ahmed S, Hodgkin J, Hengartner MO. A conserved checkpoint pathway mediates DNA damage-induced apoptosis and cell cycle arrest in C. elegans . Mol Cell. 2000;5:435–443. doi: 10.1016/S1097-2765(00)80438-4. [DOI] [PubMed] [Google Scholar]
  • 13.Blum ES, Driscoll M, Shaham S. Noncanonical cell death programs in the nematode Caenorhabditis elegans . Cell Death Differ. 2008;15:1124–1131. doi: 10.1038/cdd.2008.56. [DOI] [PubMed] [Google Scholar]
  • 14.Chung S, Gumienny TL, Hengartner MO, Driscoll M. A common set of engulfment genes mediates removal of both apoptotic and necrotic cell corpses in C. elegans . Nat Cell Biol. 2000;2:931–937. doi: 10.1038/35046585. [DOI] [PubMed] [Google Scholar]
  • 15.Fraser AG, McCarthy NJ, Evan GI. drICE is an essential caspase required for apoptotic activity in Drosophila cells. EMBO J. 1997;16:6192–6199. doi: 10.1093/emboj/16.20.6192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Berry DL, Baehrecke EH. Growth arrest and autophagy are required for salivary gland cell degradation in Drosophila . Cell. 2007;131:1137–1148. doi: 10.1016/j.cell.2007.10.048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Nicholson DW. Caspase structure, proteolytic substrates, and function during apoptotic cell death. Cell Death Differ. 1999;6:1028–1042. doi: 10.1038/sj.cdd.4400598. [DOI] [PubMed] [Google Scholar]
  • 18.Salvesen GS, Dixit VM. Caspase activation: the induced-proximity model. Proc Natl Acad Sci USA. 1999;96:10964–10967. doi: 10.1073/pnas.96.20.10964. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Boatright KM, Renatus M, Scott FL, Sperandino 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]
  • 20.Riedl SJ, Shi Y. Molecular mechanisms of caspase regulation during apoptosis. Nat Rev Mol Cell Biol. 2004;5:897–907. doi: 10.1038/nrm1496. [DOI] [PubMed] [Google Scholar]
  • 21.Luthi AU, Martin SJ. The CASBAH: a searchable database of caspase substrates. Cell Death Differ. 2007;14:641–650. doi: 10.1038/sj.cdd.4402103. [DOI] [PubMed] [Google Scholar]
  • 22.Ashkenazi A, Dixit VM. Death receptors: signaling and modulation. Science. 1998;281:1305–1308. doi: 10.1126/science.281.5381.1305. [DOI] [PubMed] [Google Scholar]
  • 23.Lavrik I, Golks A, Krammer PH. Death receptor signaling. J Cell Sci. 2005;118:265–267. doi: 10.1242/jcs.01610. [DOI] [PubMed] [Google Scholar]
  • 24.Borner C. The Bcl-2 protein family: sensors and checkpoints for life-or-death decisions. Mol Immunol. 2003;39:615–647. doi: 10.1016/S0161-5890(02)00252-3. [DOI] [PubMed] [Google Scholar]
  • 25.Youle RJ, Strasser A. The Bcl-2 protein family: opposing activities that mediate cell death. Nat Rev Mol Cell Biol. 2008;9:47–59. doi: 10.1038/nrm2308. [DOI] [PubMed] [Google Scholar]
  • 26.Puthalakath H, Strasser A. Keeping killers on a tight leash: transcriptional and post-translational control of the pro-apoptotic activity of BH3-only proteins. Cell Death Differ. 2002;9:505–512. doi: 10.1038/sj.cdd.4400998. [DOI] [PubMed] [Google Scholar]
  • 27.Wang X. The expanding role of mitochondria in apoptosis. Genes Dev. 2001;15:2922–2933. [PubMed] [Google Scholar]
  • 28.Li P, Nijhawan D, Budihardjo I, Srinivasula SM, Ahmad M, Alnemri ES, Wang X. Cytochrome c and dATP-dependent formation of Apaf-1/caspase-9 complex initiates an apoptotic protease cascade. Cell. 1997;91:479–489. doi: 10.1016/S0092-8674(00)80434-1. [DOI] [PubMed] [Google Scholar]
  • 29.Lindsten T, Ross AJ, King A, Zong WX, Rathmell JC, Shiels HA, Ulrich E, Waymire KG, Mahar P, Frauwirth K, Chen Y, Wei M, Eng VM, Adelman DM, Simon MC, Ma A, Golden JA, Evan G, Korsmeyer SJ, MacGregor GR, Thompson CB. The combined functions of proapoptotic Bcl-2 family members BAX and BAK are essential for normal development of multiple tissues. Mol Cell. 2000;6:1389–1399. doi: 10.1016/S1097-2765(00)00136-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Wei MC, Zong WX, Cheng EH, Lindsten T, Panoutsakopoulou V, Ross AJ, Roth KA, MacGregor GR, Thompson CB, Korsmeyer SJ. Proapoptotic BAX and BAK: a requisite gateway to mitochondrial dysfunction and death. Science. 2001;292:727–730. doi: 10.1126/science.1059108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Salmena L, Lemmers B, Hakem A, Matysiak-Zablocki E, Murakami K, Au PY, Berry DM, Tamblyn L, Shehabeldin A, Migon E, Wakeham A, Bouchard D, Yeh WC, McGlade JC, Ohashi PS, Hakem R. Essential role for caspase 8 in T-cell homeostasis and T-cell-mediated immunity. Genes Dev. 2003;17:883–895. doi: 10.1101/gad.1063703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.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 TNR receptors, Fas/Apo1, and DR3 and is lethal perinatally. Immunity. 1998;9:267–276. doi: 10.1016/S1074-7613(00)80609-3. [DOI] [PubMed] [Google Scholar]
  • 33.Grzela T, Krauze A, Grzela K, Lazarczyk M, Niderla J, Brawura-Biskupski-Samaha R, Dziunycz P, Milewski L, Korczak-Kowalska G, Kulus M. Impaired apoptosis of lymphocytes derived from patient with decreased expression of caspase-8 results in Alps-like phenotype. Int J Mol Med. 2004;14:937–942. [PubMed] [Google Scholar]
  • 34.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]
  • 35.Straus SE, Sneller M, Lenardo MJ, Puck JM, Strober W. An inherited disorder of lymphocyte apoptosis: the autoimmune lymphoproliferative syndrome. Ann Intern Med. 1999;130:591–601. doi: 10.7326/0003-4819-130-7-199904060-00020. [DOI] [PubMed] [Google Scholar]
  • 36.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]
  • 37.Cauwels A, Janssen B, Waeytens A, Cuvelier C, Brouckaert P. Caspase inhibition causes hyperacute tumor necrosis factor-induced shock via oxidative stress and phospholipase A2. Nat Immunol. 2003;4:387–393. doi: 10.1038/ni914. [DOI] [PubMed] [Google Scholar]
  • 38.Holler N, Zaru R, Micheau O, Thome M, Attinger A, Valitutti S, Bodmer JL, Schneider P, Seed B, Tschopp J. Fas triggers an alternative, caspase-8 independent cell death pathway using the kinase RIP as effector molecule. Nat Immunol. 2000;1:489–495. doi: 10.1038/82732. [DOI] [PubMed] [Google Scholar]
  • 39.He E, Wang L, Miao L, Wang T, Du F, Zhao L, Wang X. Receptor interacting protein kinase-3 determines cellular necrotic response to TNF-α. Cell. 2009;137:1100–1111. doi: 10.1016/j.cell.2009.05.021. [DOI] [PubMed] [Google Scholar]
  • 40.Hakem R, Hakem A, Duncan GS, Henderson JT, Woo M, Soengas MS, Elia A, de la Pompa JL, Kagi D, Khoo W, Potter J, Yoshida R, Kaufman SA, Lowe SW, Penninger JM, Mak TW. Differential requirement for caspase-9 in apoptotic pathways in vivo. Cell. 1998;94:339–352. doi: 10.1016/S0092-8674(00)81477-4. [DOI] [PubMed] [Google Scholar]
  • 41.Kuida K, Haydar TF, Kuan CY, Gu Y, Taya C, Karasuyama H, Su MS, Rakic P, Flavell RA. Reduced apoptosis and cytochrome c-mediated caspase activation in mice lacking caspase-9. Cell. 1998;94:325–337. doi: 10.1016/S0092-8674(00)81476-2. [DOI] [PubMed] [Google Scholar]
  • 42.Kuida K, Zheng TS, Na S, Kuan C, Yang D, Karasuyama H, Rakic P, Flavell RA. Decreased apoptosis in the brain and premature lethality in CPP32-deficient mice. Nature. 1996;384:368–372. doi: 10.1038/384368a0. [DOI] [PubMed] [Google Scholar]
  • 43.Lakhani SA, Masud A, Kuida A, Porter GA, Jr, Booth CJ, Mehal WZ, Inayat I, Flavell RA. Caspases 3 and 7: key mediators of mitochondrial events of apoptosis. Science. 2006;311:847–851. doi: 10.1126/science.1115035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Du C, Fang M, Li X, Li L, Wang X. Smac, a mitochondrial protein that promotes cytochrome c-dependent caspase activation by eliminating IAP inhibition. Cell. 2000;102:33–42. doi: 10.1016/S0092-8674(00)00008-8. [DOI] [PubMed] [Google Scholar]
  • 45.Verhagen AM, Ekert PG, Pakusch M, Silke J, Connolly LM, Reid GE, Moritz RL, Simpson RJ, Vaux DL. Identification of DIABLO, a mammalian protein that promotes apoptosis by binding to and antagonizing IAP proteins. Cell. 2000;102:43–53. doi: 10.1016/S0092-8674(00)00009-X. [DOI] [PubMed] [Google Scholar]
  • 46.Suzuki Y, Imai Y, Nakayama H, Takahashi K, Takio K, Takahashi R. A serine protease HtrA2, is released from the mitochondria and interacts with XIAP, inducing cell death. Mol Cell. 2001;8:613–621. doi: 10.1016/s1097-2765(01)00341-0. [DOI] [PubMed] [Google Scholar]
  • 47.Verhagen AM, Silke J, Ekert PG, Pakusch M, Kaufmann H, Connolly LM, Day CL, Tikoo A, Burke R, Wrobel C, Moritz RL, Simpson RJ, Vaux DL. HtrA2 promotes cell death through its serine protease activity and its ability to antagonize inhibitor of apoptosis proteins. J Biol Chem. 2002;277:445–454. doi: 10.1074/jbc.M109891200. [DOI] [PubMed] [Google Scholar]
  • 48.Li LY, Luo X, Wang X. Endonuclease G is an apoptotic DNase when released from mitochondria. Nature. 2001;412:95–99. doi: 10.1038/35083620. [DOI] [PubMed] [Google Scholar]
  • 49.Candé C, Vahsen N, Kouranti I, Schmitt E, Daugas E, Spahr C, Luban J, Kroemer RT, Giordanetto F, Garrido C, Penninger JM, Kroemer G. AIF and cyclophilin A cooperate in apoptosis-associated chromatinolysis. Oncogene. 2004;23:1514–1521. doi: 10.1038/sj.onc.1207279. [DOI] [PubMed] [Google Scholar]
  • 50.Galluzzi L, Joza N, Tasdemir E, Maiuri MC, Hengartner M, Abrams JM, Tavernarakis N, Penninger J, Madeo F, Kroemer G. No death without life: vital functions of apoptotic effectors. Cell Death Differ. 2008;15:1113–1123. doi: 10.1038/cdd.2008.28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.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]
  • 52.Martinon F, Burns K, Tschopp J. The inflammasome: a molecular platform triggering activation of inflammatory caspases and processing of pro-IL-1 beta. Mol Cell. 2002;10:417–426. doi: 10.1016/S1097-2765(02)00599-3. [DOI] [PubMed] [Google Scholar]
  • 53.Wang S, Miura M, Jung YK, Zhu H, Li E, Yuan J. Murine caspase-11, an ICE-interacting protease, is essential for the activation of ICE. Cell. 1998;92:501–509. doi: 10.1016/S0092-8674(00)80943-5. [DOI] [PubMed] [Google Scholar]
  • 54.Li P, Allen H, Banerjee S, Franklin S, Herzog L, Johnston C, McDowell J, Paskind M, Rodman L, Salfeld J, Towne E, Tracey D, Wardwell S, Feng-Yi W, Wong W, Kamen R, Seshadri T. Mice deficient in IL-1 beta-converting enzyme are defective in production of mature IL-1 beta and resistant to endotoxic shock. Cell. 1995;80:401–411. doi: 10.1016/0092-8674(95)90490-5. [DOI] [PubMed] [Google Scholar]
  • 55.Bergsbaken T, Fink SL, Cookson BT. Pyroptosis: host cell death and inflammation. Nat Rev Microbiol. 2009;7:99–109. doi: 10.1038/nrmicro2070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.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]
  • 57.Denecker G, Hoste E, Gilbert B, Hochepied T, Ovaere P, Lippens S, Van den Broecke C, Van Damme P, D’Herde K, Hachem JP, Borgonie G, Presland RB, Schoonjans L, Libert C, Vandekerckhove J, Gevaert K, Vandenabeele P, Declercq W. Caspase-14 protects against epidermal UVB photodamage and water loss. Nat Cell Biol. 2007;9:666–674. doi: 10.1038/ncb1597. [DOI] [PubMed] [Google Scholar]
  • 58.Wang L, Miura M, Bergeron L, Zhu H, Yuan J. Ich-1, an Ice/ced-3-related gene, encodes both positive and negative regulators of programmed cell death. Cell. 1994;78:739–750. doi: 10.1016/S0092-8674(94)90422-7. [DOI] [PubMed] [Google Scholar]
  • 59.Bergeron L, Perez GI, MacDonald G, Shi L, Sun Y, Jurisicova A, Varmuza S, Latham KE, Flaws JA, Salter JC, Hara H, Moskowitz MA, Li E, Greenberg A, Tilly JL, Yuan J. Defects in regulation of apoptosis in caspase-2-deficient mice. Genes Dev. 1998;12:1304–1314. doi: 10.1101/gad.12.9.1304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.O’Reilly LA, Ekert PG, Harvey N, Marsden V, Cullen L, Vaux DL, Hacker G, Magnusson C, Pakusch M, Cecconi F, Kuida K, Strasser A, Huang DC, Kumar S. Caspase-2 is not required for thymocyte or neuronal apoptosis even through cleavage of caspase-2 is dependent on both Apaf-1 and caspase-9. Cell Death Differ. 2002;9:832–841. doi: 10.1038/sj.cdd.4401033. [DOI] [PubMed] [Google Scholar]
  • 61.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]
  • 62.Read SH, Baliga BC, Ekert PG, Vaux DL, Kumar S. A novel Apaf-1-independent putative caspase-2 activation complex. J Cell Biol. 2002;159:739–745. doi: 10.1083/jcb.200209004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Shi M, Vivian CJ, Lee K-J, 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. DNA-PKcs-PIDDosome: a nuclear caspase-2-activating complex with role in G2/M checkpoint maintenance. Cell. 2009;136:508–520. doi: 10.1016/j.cell.2008.12.021. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 64.Guo Y, Srinivasula SM, Druilhe A, Fernandes-Alnemri T, Alnemri ES. Caspase-2 induces apoptosis by releasing proapoptotic proteins from mitochondria. J Biol Chem. 2002;277:13430–13437. doi: 10.1074/jbc.M108029200. [DOI] [PubMed] [Google Scholar]
  • 65.Marsden VS, Ekert PG, Van Delft M, Vaux DL, Adams JM, Strasser A. Bcl-2-regulated apoptosis and cytochrome c release can occur independently of both caspase-2 and caspase-9. J Cell Biol. 2004;165:775–780. doi: 10.1083/jcb.200312030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Slee EA, Harte MT, Kluck RM, Wolf BB, Casiano CA, Newmeyer DD, Wang HG, Reed JC, Nicholson DW, Alnemri ES, Green DR, Martin SJ. Ordering of cytochrome c-initiated caspase cascade: hierarchical activation of caspases-2, -3, -6, -7, -8 and -10 in a caspase-9-dependent manner. J Cell Biol. 1999;144:281–292. doi: 10.1083/jcb.144.2.281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Ruchaud S, Korfali N, Villa P, Kottke TJ, Dingwall C, Kaufmann SH, Earnshaw WC. Caspase-6 gene disruption reveals a requirement for lamin A cleavage in apoptotic chromatin condensation. EMBO J. 2002;21:1967–1977. doi: 10.1093/emboj/21.8.1967. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Slee EA, Adrain C, Martin SJ. Executioner caspase-3, -6, and -7 perform distinct, non-redundant roles during the demolition phase of apoptosis. J Biol Chem. 2001;276:7320–7326. doi: 10.1074/jbc.M008363200. [DOI] [PubMed] [Google Scholar]
  • 69.Cowling V, Downward J. Caspase-6 is the direct activator of caspase-8 in the cytochrome c-induced apoptosis signalling 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]
  • 70.O’Connell AR, Stenson-Cox C. A more serine way to die: defining the characteristics of serine protease-mediated cell death cascades. Biochem Biophys Acta. 2007;1773:1491–1499. doi: 10.1016/j.bbamcr.2007.08.002. [DOI] [PubMed] [Google Scholar]
  • 71.Moffitt KL, Martin SL, Walker B. The emerging role of serine proteases in apoptosis. Biochem Soc Trans. 2007;35:559–560. doi: 10.1042/BST0350559. [DOI] [PubMed] [Google Scholar]
  • 72.Conus S, Simon H-U. Cathepsins: key modulators of cell death and inflammatory responses. Biochem Pharmacol. 2008;76:1374–1382. doi: 10.1016/j.bcp.2008.07.041. [DOI] [PubMed] [Google Scholar]
  • 73.Boya P, Kroemer G. Lysosomal membrane permeabilzation in cell death. Oncogene. 2008;27:6434–6451. doi: 10.1038/onc.2008.310. [DOI] [PubMed] [Google Scholar]
  • 74.Ruggiero V, Johnson SE, Baglioni C. Protection from tumor necrosis factor cytotoxicity by protease inhibitors. Cell Immunol. 1987;107:317–325. doi: 10.1016/0008-8749(87)90240-1. [DOI] [PubMed] [Google Scholar]
  • 75.Williams MS, Henkart PA. Apoptotic cell death induced by intracellular proteolysis. J Immunol. 1994;153:4247–4255. [PubMed] [Google Scholar]
  • 76.Abate A, Schroder H. Protease inhibitors protect macrophages from lipopolysaccharide-induced cytotoxicity: possible role for NF-kappaB. Life Sci. 1998;62:1081–1088. doi: 10.1016/S0024-3205(98)00031-9. [DOI] [PubMed] [Google Scholar]
  • 77.Masson D, Tschopp J. A family of serine esterases in lytic granules of cytolytic T lymphocytes. Cell. 1987;49:679–685. doi: 10.1016/0092-8674(87)90544-7. [DOI] [PubMed] [Google Scholar]
  • 78.Shi L, Kam CM, Powers JC, Aebersold R, Greenberg AH. Purification of three cytotoxic lymphocyte granule serine proteases that induce apoptosis through distinct substrate and target cell interactions. J Exp Med. 1992;176:1521–1529. doi: 10.1084/jem.176.6.1521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Chowdhury D, Lieberman J. Death by a thousand cuts: granzyme pathways of programmed cell death. Annu Rev Immunol. 2008;26:389–420. doi: 10.1146/annurev.immunol.26.021607.090404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Chen C, Darrow AL, Qi JS, D’Andrea MR, Andrade-Gordon P. A novel serine protease predominately expressed in macrophages. Biochem J. 2003;374:97–107. doi: 10.1042/BJ20030242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.De Bruin EC, Meersma D, De Wilde J, Den Otter I, Schipper EM, Medema JP, Peltenburg LT. A serine protease is involved in the initiation of DNA damage-induced apoptosis. Cell Death Differ. 2003;10:1204–1212. doi: 10.1038/sj.cdd.4401296. [DOI] [PubMed] [Google Scholar]
  • 82.Strasser A, Jost PJ, Nagata S. The many roles of Fas receptor signaling in the immune system. Immunity. 2009;30:180–192. doi: 10.1016/j.immuni.2009.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Trapani JA, Smyth MJ. Functional significance of the perforin/granzyme cell death pathway. Nat Rev Immunol. 2002;2:735–747. doi: 10.1038/nri911. [DOI] [PubMed] [Google Scholar]
  • 84.Pardo J, Bosque A, Brehm R, Wallich R, Naval J, Mullbacher A, Anel A, Simon MM. Apoptotic pathways are selectively activated by granzyme A and/or granzyme B in CTL-mediated target cell lysis. J Cell Biol. 2004;167:457–468. doi: 10.1083/jcb.200406115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Martin SJ, Amarante-Mendes GP, Shi L, Chuang TH, Casiano CA, O’Brien CA, Fitzgerald P, Tan EM, Bokoch GM, Greenberg AH, Green DR. The cytotoxic cell protease granzyme B initiates apoptosis in a cell-free system by proteolytic processing and activation of the ICE/CED-3 family protease, CPP32, via a novel two-step mechanism. EMBO J. 1996;15:2407–2416. [PMC free article] [PubMed] [Google Scholar]
  • 86.Barry M, Heibein JA, Pinkoski MJ, Lee SF, Moyer RW, Green DR, Bleackley RC. Granzyme B short-circuits the need for caspase 8 activity during granule-mediated cytotoxic T-lymphocyte killing by directly cleaving Bid. Mol Cell Biol. 2000;20:3781–3794. doi: 10.1128/MCB.20.11.3781-3794.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Waterhouse NJ, Sedelies KA, Trapani JA. Role of Bid-induced mitochondrial outer membrane permeabilization in granzyme B-induced apoptosis. Cell Death Differ. 2006;7:17–24. doi: 10.1111/j.1440-1711.2005.01416.x. [DOI] [PubMed] [Google Scholar]
  • 88.Han J, Goldstein LA, Gastman BR, Froelich CJ, Xin XM, Rabinowich H. Disruption of Mcl-1 by granzyme B: implications for Bim-mediated mitochondrial apoptotic event. J BiolChem. 2005;279:22020–22029. doi: 10.1074/jbc.M313234200. [DOI] [PubMed] [Google Scholar]
  • 89.Martinvalet D, Zhu P, Lieberman J. Granzyme A induces caspase-independent mitochondrial damage, a required first step for apoptosis. Immunity. 2005;22:355–370. doi: 10.1016/j.immuni.2005.02.004. [DOI] [PubMed] [Google Scholar]
  • 90.Fan Z, Beresford PJ, Oh DY, Zhang D, Lieberman J. Tumor suppressor NM23-H1 is a granzyme A-activated DNase during CTL-mediated apoptosis, and the nucleosome assembly protein SET is its inhibitor. Cell. 2003;112:659–672. doi: 10.1016/S0092-8674(03)00150-8. [DOI] [PubMed] [Google Scholar]
  • 91.Metkar SS, Menaa C, Pardo J, Wang B, Wallich R, Freudenberg M, Kim S, Raja SM, Shi L, Simon MM, Froelich CJ. Human and mouse granzyme A induce a pro-inflammatory cytokine response. Immunity. 2008;29:720–733. doi: 10.1016/j.immuni.2008.08.014. [DOI] [PubMed] [Google Scholar]
  • 92.Martins LM, Morrison A, Klupsch K, Fedele V, Moisoi N, Teismann P, Abuin A, Grau E, Geppert M, Livi GP, Creasy CL, Martin A, Hargreaves I, Heales SJ, Okada H, Brandner S, Schulz JB, Mak T, Downward J. Neuroprotective role of the reaper-related serine protease HtrA2/Omi revealed by targeted deletion in mice. Mol Cell Biol. 2004;24:9848–9862. doi: 10.1128/MCB.24.22.9848-9862.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Chao JR, Parganas E, Boyd K, Hong CY, Opferman JT, Ihle JN. Hax1-mediated processing of HtrA2 by Parl allows survival of lymphocytes and neurons. Nature. 2008;452:98–102. doi: 10.1038/nature06604. [DOI] [PubMed] [Google Scholar]
  • 94.Nagata S. DNA degradation in development and programmed cell death. Annu Rev Immunol. 2007;23:853–875. doi: 10.1146/annurev.immunol.23.021704.115811. [DOI] [PubMed] [Google Scholar]
  • 95.Beresford PJ, Zhang D, Oh DY, Fan Z, Greer EL, Russo ML, Jaju M, Lieberman J. Granzyme A activates an endoplasmic reticulum-associated caspase-independent nuclease to induce single-stranded DNA nicks. J Biol Chem. 2001;276:43285–43293. doi: 10.1074/jbc.M108137200. [DOI] [PubMed] [Google Scholar]
  • 96.Dong Z, Saikumar P, Weinberg JM, Venkatachalam MA. Internucleosomal DNA cleavage triggered by plasma membrane damage during necrotic cell death. Involvement of serine but not cysteine proteases. Am J Pathol. 1997;151:1205–1213. [PMC free article] [PubMed] [Google Scholar]
  • 97.Schlegel J, Peters I, Orrenius S. Isolation and partial characterization of a protease involved in Fas-induced apoptosis. FEBS Lett. 1995;364:139–142. doi: 10.1016/0014-5793(95)00374-I. [DOI] [PubMed] [Google Scholar]
  • 98.McGrath LB, Onnis V, Campiani G, Williams DC, Zisterer DM, McGee MM. Caspase-activated DNase (CAD)-independent oligonucleosomal DNA fragmentation in chronic myeloid leukemia cells; a requirement for serine protease and Mn(2+)-dependent acidic endonuclease activity. Apoptosis. 2006;11:1473–1487. doi: 10.1007/s10495-006-8968-4. [DOI] [PubMed] [Google Scholar]
  • 99.Altairac S, Wright SC, Courtois Y, Torriglia A. l-DNase II activation by the 24 kD apoptotic protease (AP24) in TNF alpha-induced apoptoiss. Cell Death Differ. 2003;10:1109–1111. doi: 10.1038/sj.cdd.4401293. [DOI] [PubMed] [Google Scholar]
  • 100.Wright SC, Wei QS, Kinder DH, Larrick JW. Biochemical pathways of apoptosis: nicotinamide adenine dinucleotide-deficient cells are resistant to tumor necrosis factor or ultraviolet light activation of the 24-kD apoptotic protease and DNA fragmentation. J Exp Med. 1996;183:463–471. doi: 10.1084/jem.183.2.463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Wright SC, Schellenberger U, Wang H, Kinder DH, Talhouk JW, Larrick JW. Activation of CPP32-like proteases is not sufficient to trigger apoptosis: inhibition of apoptosis by agents that suppress activation of AP24, but not CPP32-like activity. J Exp Med. 1997;186:1107–1117. doi: 10.1084/jem.186.7.1107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Wright SC, Zheng H, Zhong J. Tumor cell resistance to apoptosis due to a defect in the activation of sphingomyelinase and the 24-kDa apoptotic protease (AP24) FASEB J. 1996;10:325–332. doi: 10.1096/fasebj.10.2.8641566. [DOI] [PubMed] [Google Scholar]
  • 103.Wright SC, Schellenberger U, Ji L, Wang H, Larrick JW. Calmodulin-dependent protein kinase II mediates signal transduction in apoptosis. FASEB J. 1997;11:843–849. doi: 10.1096/fasebj.11.11.9285482. [DOI] [PubMed] [Google Scholar]
  • 104.Wright SC, Wang H, Wei QS, Kinder DH, Larrick JW. Bcl-2 mediated resistance to apoptosis is associated with glutathione-induced inhibition of AP24 activation and nuclear DNA fragmentation. Cancer Res. 1998;58:5570–5576. [PubMed] [Google Scholar]
  • 105.Dong Z, Saikumar P, Patel Y, Weinberg JM, Venkatachalam MA. Serine protease inhibitors suppress cytochrome c-mediated caspase-9 activation and apoptosis during hypoxia-reoxygenation. Biochem J. 2000;347:669–677. doi: 10.1042/0264-6021:3470669. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.O’Connell AR, Holohan C, Torriglia A, Lee BW, Stenson-Cox C. Characterization of a serine protease-mediated cell death program activated in human leukemic cells. Exp Cell Res. 2006;312:27–39. doi: 10.1016/j.yexcr.2005.10.003. [DOI] [PubMed] [Google Scholar]
  • 107.O’Connell AR, Lee BW, Stenson-Cox C. Caspase-dependent activation of chymotrypsin-like proteases mediates nuclear events during Jurkat C cell apoptosis. Biochem Biophys Res Commun. 2006;345:608–616. doi: 10.1016/j.bbrc.2006.04.147. [DOI] [PubMed] [Google Scholar]
  • 108.Potempa J, Korzus E, Travis J. The serpin superfamily of proteinase inhibitors: structure, function, and regulation. J Biol Chem. 1994;269:15957–15960. [PubMed] [Google Scholar]
  • 109.Ray CA, Black RA, Kronheim SR, Greenstreet TA, Sleath PR, Salvesen GS, Pickup DJ. Viral inhibition of inflammation: cowpox virus encodes an inhibitor of the interleukin-1 beta converting enzyme. Cell. 1992;69:597–604. doi: 10.1016/0092-8674(92)90223-Y. [DOI] [PubMed] [Google Scholar]
  • 110.Srinivasula SM, Ahman T, Fernandes-Alnemri T, Litwack G, Alnemri ES. Molecular ordering of the Fas-apoptotic pathway: the Fas/APO-1 protease Mch5 is a CrmA-inhibitable protease that activates multiple CED-3/ICE-like cysteine proteases. Proc Natl Acad Sci USA. 1996;93:14486–14491. doi: 10.1073/pnas.93.25.14486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Quan LT, Caputo A, Bleackley RC, Pickup DJ, Salvesen GS. Granzyme B is inhibited by the cowpox virus serpin cytokine response modifier A. J Biol Chem. 1995;270:10377–10379. doi: 10.1074/jbc.270.52.31046. [DOI] [PubMed] [Google Scholar]
  • 112.Tewari M, Quan LT, O’Rourke K, Desnoyers S, Zeng Z, Beidler DR, Poirier GG, Salvesen GS, Dixit VM. Yama/CPP32 beta, a mammalian homolog of CED-3, is a CrmA-inhibitable protease that cleaves the death substrate poly(ADP-ribose)polymerase. Cell. 1995;81:801–809. doi: 10.1016/0092-8674(95)90541-3. [DOI] [PubMed] [Google Scholar]
  • 113.Sun J, Bird CH, Sutton V, McDonald L, Coughlin PB, De Jong TA, Trapani JA, Bird PI. A cytosolic granzyme B inhibitor related to the viral apoptotic regulator cytokine response modifier A is present in cytotoxic lymphocytes. J Biol Chem. 1996;271:27802–27809. doi: 10.1074/jbc.271.44.27802. [DOI] [PubMed] [Google Scholar]
  • 114.Ishidoh K, Kominami D. Processing and activation of lysosomal proteinases. Biol Chem. 2002;383:1827–1831. doi: 10.1515/BC.2002.206. [DOI] [PubMed] [Google Scholar]
  • 115.Turk B, Bieth JG, Bjork I, Dolen I, Turk D, Cimerman N, Kos J, Colic A, Stoka V, Turk V. Regulation of the activity of lysosomal cystein proteinases by pH-induced inactivation and/or endogenous protein inhibitors, cystatins. Biol Chem Hoppe Seyler. 1995;376:225–230. doi: 10.1515/bchm3.1995.376.4.225. [DOI] [PubMed] [Google Scholar]
  • 116.Vasiljeva O, Turk B. Dual contrasting roles of cysteine cathepsins in cancer progression: apoptosis versus tumour invasion. Biochimie. 2008;90:380–386. doi: 10.1016/j.biochi.2007.10.004. [DOI] [PubMed] [Google Scholar]
  • 117.Gottlieb RA, Nordberg J, Skowronski E, Babiol BM. Apoptosis induced in Jurkat cells by several agents is preceded by intracellular acidification. Proc Natl Acad Sci USA. 1996;93:654–658. doi: 10.1073/pnas.93.2.654. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Boya P, Andreau K, Poncet D, Zamzami N, Perfettini JL, Metivier D, Ojcius S, Jäättelä M, Kroemer G. Lysosomal membrane permeabilization induces cell death in a mitochondrion-dependent fashion. J Exp Med. 2003;197:1323–1334. doi: 10.1084/jem.20021952. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Cirman T, Oresc K, Mazovec GD, Turk V, Reed JC, Myers RM, Salvesen GS, Turk B. Selective disruption of lysosomes in HeLa cells triggers apoptosis mediated by cleavage of Bid by multiple papain-like lysosomal cathepsins. J Biol Chem. 2004;279:3578–3587. doi: 10.1074/jbc.M308347200. [DOI] [PubMed] [Google Scholar]
  • 120.Stoka V, Turk B, Schendel SL, Kim TH, Cirman T, Snipas SJ, Ellerby LM, Bredesen D, Freeze H, Abrahamson M, Bromme D, Krajewski S, Reed JC, Yin XM, Turk V, Salvesen GS. Lysosomal protease pathways to apoptosis. Cleavage of Bid, not pro-caspases, is the most likely route. J Biol Chem. 2001;276:3149–3157. doi: 10.1074/jbc.M008944200. [DOI] [PubMed] [Google Scholar]
  • 121.Li H, Zhu H, Xu CJ, Yuan J. Cleavage of BID by caspase-8 mediates the mitochondria damage in the Fas pathway of apoptosis. Cell. 1998;94:491–501. doi: 10.1016/S0092-8674(00)81590-1. [DOI] [PubMed] [Google Scholar]
  • 122.Heinrich M, Neumeyer J, Jakob M, Hallas C, Tchikov V, Winoto-Morbach S, Wickel M, Schneider-Brachert W, Trauzold A, Hethke A, Schütze S. Cathepsin D links TNF-induced acid sphingomyelinase to Bid-mediated caspase-9 and -3 activation. Cell Death Differ. 2004;11:550–563. doi: 10.1038/sj.cdd.4401382. [DOI] [PubMed] [Google Scholar]
  • 123.Houseweart MK, Vilaythong A, Yin KM, Turk B, Noebels JL, Myers RM. Apoptosis caused by cathepsins does not require Bid signaling in an in vivo model of progressive myoclonus epilepsy (EPM1) Cell Death Differ. 2003;10:1329–1335. doi: 10.1038/sj.cdd.4401309. [DOI] [PubMed] [Google Scholar]
  • 124.Droga-Mazovec G, Bojiic L, Petelin A, Ivanova S, Romih R, Repnik U, Salvesen GS, Stoka V, Turk V, Turk B. Cysteine cathepsins trigger caspase-dependent cell death through cleavage of Bid and anti-apoptotic Bcl-2 homologues. J Biol Chem. 2008;283:19140–19145. doi: 10.1074/jbc.M802513200. [DOI] [PubMed] [Google Scholar]
  • 125.Ishisaka R, Utsumi T, Kanno T, Arita K, Katunuma N, Akiyama J, Utsumi K. Participation of a cathepsin L-type protease in the activation of caspase-3. Cell Struct Funct. 1999;465:465–470. doi: 10.1247/csf.24.465. [DOI] [PubMed] [Google Scholar]
  • 126.Ishisaka R, Kanno T, Akiyama J, Yoshioka T, Utsumi K, Utsumi T. Activation of caspase-3 by lysosomal cysteine proteases and its role in 2, 2′-azobis-(2-amidinopropane)dihydrochloride (AAPH)-induced apoptosis in HL-60 cells. J Biochem. 2001;129:35–41. doi: 10.1093/oxfordjournals.jbchem.a002833. [DOI] [PubMed] [Google Scholar]
  • 127.Guicciardi ME, Bronk SF, Werneburg N, Yin XM, Gores GJ. Bid is upstream of lysosome-mediated caspase-2 activation in tumor necrosis factor a-induced hepatocyte apoptosis. Gastroenterologia. 2005;129:269–284. doi: 10.1053/j.gastro.2005.05.022. [DOI] [PubMed] [Google Scholar]
  • 128.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]
  • 129.Foghsgaard L, Wissing D, Mauch D, Lademann U, Bastholm L, Boes M, Elling F, Leist M, Jäättelä M. Cathepsin B acts as a dominant execution protease in tumor cell apoptosis induced by tumor necrosis factor. J Cell Biol. 2001;153:999–1009. doi: 10.1083/jcb.153.5.999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Guicciardi ME, Deussing J, Miyoshi H, Bronk SF, Svingen PA, Peters C, Kaufmann SH, Gores GJ. Cathepsin B contributes to TNF-alpha-mediated hepatocyte apoptosis by promoting mitochondrial release of cytochrome c . J Clin Invest. 2000;106:1127–1137. doi: 10.1172/JCI9914. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Deiss LP, Galinka H, Berissi H, Cohen O, Kimchi A. Cathepsin D protease mediates programmed cell death induced by interferon-gamma, Fas/APO-1 and TNF-alpha. EMBO J. 1996;15:3861–3870. [PMC free article] [PubMed] [Google Scholar]
  • 132.Nagaraj NS, Vigneswaran N, Zacharias W. Cathepsin B mediates TRAIL-induced apoptosis in oral cancer cells. J Cancer Res Clin Oncol. 2006;132:171–183. doi: 10.1007/s00432-005-0053-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Werneburg NW, Guicciardi ME, Yin XM, Gores GJ. TNF-α-mediated lysosomal permeabilization is FAN- and caspase-8/Bid-dependent. Am J Physiol Gastrointest Liver Physiol. 2004;287:G436–G443. doi: 10.1152/ajpgi.00019.2004. [DOI] [PubMed] [Google Scholar]
  • 134.Gyrd-Hansen M, Farkas T, Fehrenbacher N, Bastholm L, Hoyer-Hansen M, Elling F, Wallach D, Flavell R, Kroemer G, Nylandsted J, Jäättelä M. Apoptosome-independent activation of the lysosomal cell death pathway by caspase-9. Mol Cell Biol. 2006;26:7880–7891. doi: 10.1128/MCB.00716-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Vasiljeva O, Reinheckel T, Peters C, Turk D, Turk V, Turk B. Emerging roles of cysteine cathepsins in disease and their potential as drug targets. Curr Pharm Des. 2007;13:385–401. doi: 10.2174/138161207779313542. [DOI] [PubMed] [Google Scholar]
  • 136.Tobin DJ, Foitzik K, Reinheckel T, Mecklenburg L, Botchkarev VA, Peters C, Paus R. The lysosomal protease cathepsin L is an important regulator of keratinocyte and melanocyte differentiation during hair follicle morphogenesis and cycling. Am J Pathol. 2002;160:1807–1821. doi: 10.1016/S0002-9440(10)61127-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Reinheckel T, Hagemann S, Dollwet-Mack S, Martinez E, Lohmuller T, Zlatkovic G, Tobin DJ, Maas-Szabowski N, Peters C. The lysosomal cysteine protease cathepsin L regulates keratinocyte proliferation by control of growth factor recycling. J Cell Sci. 2005;118:3387–3395. doi: 10.1242/jcs.02469. [DOI] [PubMed] [Google Scholar]
  • 138.Werneburg NW, Guicciardi ME, Bronk SF, Kaufmann SH, Gores GJ. Tumor necrosis factor-related apoptosis-inducing ligand activates a lysosomal pathway of apoptosis that is regulated by Bcl-2 proteins. J Biol Chem. 2007;282:28960–28970. doi: 10.1074/jbc.M705671200. [DOI] [PubMed] [Google Scholar]
  • 139.Oberle C, Huai J, Reinheckel T, Tacke M, Rassner M, Ekert PG, Buellesbach J, Borner C (2009) Lysosomal membrane permeabilization and cathepsin release is a Bax/Bak-dependent amplifying event of apoptosis in fibroblasts and monocytes. Cell Death Differ. doi:10.1038/cdd.2009.214 [DOI] [PubMed]

Articles from Cellular and Molecular Life Sciences: CMLS are provided here courtesy of Springer

RESOURCES