<<

Leukemia (2000) 14, 1695–1703  2000 Macmillan Publishers Ltd All rights reserved 0887-6924/00 $15.00 www.nature.com/leu REVIEW

Noncaspase in DE Johnson

Departments of Medicine and Pharmacology, University of Pittsburgh, and the University of Pittsburgh Institute, Pittsburgh, PA, USA

Biochemical and genetic analysis of apoptosis has determined appear to be important for apoptosis execution in different that intracellular proteases are key effectors of death path- cell types. ways. In particular, early studies have pointed to the primacy of proteases as mediators of execution. More recently, While the importance of in apoptosis is clearly however, evidence has accumulated that noncaspases, includ- established, recent studies have indicated that several other ing , , , and the com- types of proteases also may play a role in the execution pro- plex, also have roles in mediating and promoting . cess. The involvement of noncaspase proteases is suggested An important goal is to understand the importance of distinct by observations that inhibition of caspase proteases generally noncaspases in various forms of apoptosis, and to determine causes delays, but does not fully block, cell death resulting whether pathways mediated by noncaspase proteases inter- sect with those mediated by caspases. In this review the roles from most apoptotic stimuli. In addition, implicated noncasp- of noncaspase proteases in the biochemistry of apoptosis will ases frequently are upregulated or activated during apoptosis, be discussed. Leukemia (2000) 14, 1695–1703. and their inhibition, as with caspase inhibition, can serve to Keywords: apoptosis; B; ; ; delay apoptosis. The noncaspase proteases that have been A; granzyme B most closely linked with apoptosis are cathepsins, calpains, granzymes, and the proteasome. This review will focus on the current state of knowledge regarding the involvement and role Introduction of cathepsins, calpains, and granzymes in the execution pro- cess. The role of the proteasome complex, which has been The demise of cells via apoptosis hinges on the activation of strongly implicated in apoptosis, has been recently reviewed cellular proteases. Elucidation of the specific proteases elsewhere.6 involved in apoptotic execution began in the early 1990s with genetic and biochemical studies of apoptotic cell death in the C. elegans. These studies revealed that apoptosis in Cathepsins and apoptosis C. elegans was dependent on an intracellular bearing considerable homology to the human interleukin-1β con- The cathepsin protease family consists of at least 12 known verting , or ICE.1–3 Subsequent studies have led to the members.7,8 Cathepsins can be subdivided into three distinct identification of 13 mammalian proteases that are related to groups, based on the that comprises the ICE, and these proteases, along with ICE, have been termed residue: (1) serine proteases (cathepsins A and G), (2) cysteine caspase proteases (caspase-1 to caspase-13).4,5 The term cas- proteases (cathepsins B, C, H, K, L, S, and T), and (3) aspartate pase, or c-aspase, reflects the fact that these are cyst- proteases (cathepsins D and E). Like the caspases, cathepsins eine proteases that cleave substrate proteins after aspartate are synthesized as inactive zymogens, and activation involves residues. Caspases are initially synthesized in the cell as proteolytic processing.9–13 As discussed below, the involve- inactive zymogens. However, in response to an apoptotic ment of cathepsins in apoptotic cell death has primarily been stimulus, initiator caspases such as caspase-8 or -9 undergo studied in nonhematopoietic systems. Evidence from these processing to active forms.5 Active initiator caspases then systems, however, should provide the basis for investigating cleave and activate downstream executioner caspases such as the role of cathepsins in the death of both normal and malig- caspase-3 or -7, thereby initiating a cascade of caspase acti- nant hematopoietic cells.14 vation. Once activated, executioner caspases cleave cellular The most extensive evidence linking cathepsins with substrate proteins promoting the destruction of the cell. apoptosis has come from studies of the , The role of caspases in mammalian apoptosis has been , and the aspartate protease, cathepsin D. There- intensively investigated using a variety of approaches: (1) fore, this review will focus on these two members of the pharmacologic inhibition of caspases using inhibitors; cathepsin family. Both cathepsin B and cathepsin D are found (2) inhibition of caspases with endogenous or viral inhibitory primarily in or endosomes. Historically, it has been proteins; and (3) targeted disruption. These studies have presumed that these proteases are mainly involved in the ter- shown that inhibition of caspases significantly delays minal degradation of proteins within the lysosomal compart- apoptosis induced by a wide variety of stimuli in a variety of ment. However, both proteases can also be secreted, and different cell types. Moreover, gene knockout experiments once secreted can degrade collagen, fibronectin, laminin, and have shown that different members of the caspase family proteoglycans.9,15 The degradation of components by cathepsins B and D accounts for the ability of these proteases to promote or malignant invasion.16–19 Indeed, overexpression and secretion of cathep- Correspondence: DE Johnson, Division of Hematology/Oncology, sin D, which can be induced by estrogen, closely correlates University of Pittsburgh, Biomedical Science Tower, Room BST 20–23 E1055, 211 Lothrop Street, Pittsburgh, PA, 15213–2582, USA; Fax: with high metastatic potential in breast cancer. Additional (412) 624-7794 evidence (described below) suggests that cathepsins B and D Received 20 April 2000; accepted 22 May 2000 are translocated to the cytoplasm during apoptosis. This is sig- Review DE Johnson 1696 nificant, since most of the known apoptosis execution path- were also associated with induction and processing of cathep- ways occur in the cytoplasm. Thus, the translocation of sin D protein. cathepsins to the cytoplasm may allow these proteases to Cathepsin D also participates in the execution of cells intersect with and augment other apoptosis signaling mech- treated with chemotherapy drugs or radiation. Wu et al,40 anisms. using a subtractive hybridization strategy, identified cathepsin Early evidence suggesting a role for cathepsins B and D in D mRNA as an upregulated transcript in doxorubicin-treated apoptosis came from studies of apoptotic cell death in rat cells. Induction of cathepsin D protein was observed in ML1 prostate and mammary tissues. Following androgen with- leukemic cells treated with doxorubicin, etoposide, or γ- drawal in prostate, or following lactation in breast, massive irradiation. Upregulation of cathepsin D may be impacted by apoptosis occurs in the epithelial cells of these tissues.24,25 p53, as two p53 binding sites were found in the promoter Concurrent with this apoptosis, there is upregulation of mRNA region of cathepsin D. Wu et al40 further showed that drug- and protein for both cathepsin B and D.26,27 While these initial induced apoptosis was inhibited by pepstatin A. Moreover, findings suggested a role for cathepsins in regressing prostate fibroblasts derived from cathepsin D−/− gene knockout mice and mammary gland, they did not directly demonstrate displayed enhanced resistance to adriamycin and etoposide cathepsin involvement in the execution pathways. It remained relative to fibroblasts from wild-type mice.40 Similar studies possible that cathepsins were simply needed for ‘cleanup’ in by Roberts et al29 have suggested the involvement of both the dying cells. cathepsin B and cathepsin D in camptothecin-induced More definitive experiments showing a role for cathepsins apoptosis of a hepatocyte cell line (Hep3B). Following treat- in apoptotic execution have come from studies of bile salt- ment of Hep3B cells with camptothecin, an increase in the induced apoptosis in cultured hepatocytes. This in vitro model cellular activity of cathepsin B was observed, as measured by mimics apoptosis that occurs in human hepatocytes in dis- cleavage of z-ValLeuLys-7-amino-4-chloromethylcoumarin. eases where normal bile flow is impaired. In cell culture, bile Increased activity of cathepsin D, as measured with the sub- salt-induced apoptosis was found to be markedly inhibited by strate D-PheSerPhePheAlaAla-p-aminobenzoate, was also CA-074-Me, a specific inhibitor of cathepsin B, and by pepsta- detected. Inhibition of cathepsins B and D with CA-074-Me tin A, an inhibitor of cathepsin D.28,29 Antisense-mediated and pepstatin A, respectively, markedly diminished downregulation of cathepsin B also markedly inhibited hepa- camptothecin-induced apoptosis. Also, as observed in bile tocyte apoptosis following treatment with bile salts.28 salt-induced apoptosis of hepatocytes, cathepsin D is Together, these findings showed that, similar to caspases, upstream of cathepsin B in the camptothecin-induced cell cathepsins B and D, can be important components of death pathway. apoptosis execution pathways. Interestingly, pepstatin A was While inhibition of cathepsins clearly delays some forms of found to inhibit bile salt-induced cathepsin B activation, while apoptosis, any discussion of the role of cathepsins in apoptotic CA-074-Me failed to impact cathepsin D activation.29 This execution should consider the issue of subcellular localiz- revealed that during bile salt-induced apoptosis, cathepsin D ation. Apoptosis pathways have been is activated upstream of cathepsin B. Thus, like the caspases, found to occur in the cytoplasm, on the inner surface of the cathepsins may be activated in a cascade-like fashion during plasma membrane, in mitochondria, and in the nucleus.5,41–46 apoptosis. In support of this idea, cathepsin D has been shown In particular, caspase-mediated signaling occurs predomi- to directly cleave and activate cathepsin B.30,31 nantly in the cytoplasm.5,47,48 By contrast, cathepsins B and Cathepsins B and D exert opposing effects on apoptosis in D are localized in lysosomes, or are secreted by the cell. This serum- and neurotrophic factor-deprived neuronal cells. Dur- raises the question as to how these enzymes can facilitate ing apoptosis of serum-deprived rat PC12 cells, the levels of apoptosis signaling. An emerging line of evidence suggests cathepsin B protein decline while those of cathepsin D that during apoptosis, cathepsins B and D are translocated increase.32 Interestingly, cathepsin D levels are also elevated from lysosomes to other subcellular localizations. Three stud- in the pyramidal of Alzheimers patients.33 In PC12 ies have utilized gold-labeled antibodies followed by electron cells, inhibition of cathepsin B with CA-074, or downregul- microscopic immunocytochemistry to examine cathepsin D ation of cathepsin B with antisense oligonucleotides resulted localization during apoptosis.49–51 Li et al50 found that treat- in enhanced apoptosis.34 By contrast, inhibition of cathepsin ment of the mouse macrophage cell line J-774 or human D with pepstatin A significantly inhibited PC12 cell apoptosis. macrophages with apoptosis-inducing oxidized LDL changed Similar results were seen using dorsal root ganglion neurons the pattern of cathepsin D staining from granular lysosomal to deprived of nerve growth factor.34 It will be interesting to see diffuse cytoplasmic. Similar results were seen in rat myocytes whether these findings can be extended to hematopoietic treated with the quinone naphthazarin,51 and in cells treated cells. Many hematopoietic cells are dependent on specific with hydrogen peroxide.49 In the case of bile salt-induced for survival, and undergo apoptosis when deprived apoptosis of hepatocytes, Roberts et al28 performed subcellu- of these essential factors.35,36 Future studies are needed to lar fractionation, followed by Western blotting, and found that define the role and involvement of cathepsins during preexisting cathepsin B redistributed to the nucleus in treated withdrawal-induced apoptosis.37,38 cells. This group also expressed a cathepsin B-green fluor- The involvement of cathepsin D in the execution pathways escent protein chimeric molecule, and observed translocation triggered by a variety of other stimuli has been demonstrated of the fluorescent signal to the hepatocyte nucleus following by Deiss et al.39 This group isolated antisense cathepsin D treatment with bile salts. Another group has examined the cDNA while screening an antisense library for clones that localization of cathepsin B in response to the agent atractylo- could inhibit IFN-γ-induced apoptosis in HeLa cells. In side.52 Atractyloside is known to induce mitochondrial per- addition to apoptosis caused by IFN-γ, antisense cathepsin D meability transition, and subsequent apoptosis. Surprisingly, it also markedly inhibited Fas-induced apoptosis. Moreover, was shown that atractyloside caused the release of cathepsin pepstatin A was found to inhibit both forms of cell death, as B from highly purified lysosomes. Thus, current evidence indi- well as apoptosis induced by TNF-α treatment of U937 histi- cates that cathepsins may be released from lysosomal com- ocytic lymphoma cells. IFN-γ- and TNF-α-induced apoptosis partments during some forms of apoptosis. It remains unclear,

Leukemia Review DE Johnson 1697 however, whether this is a general apoptosis phenomenon, or Another interesting feature of the calpains is their ability to is restricted to execution induced by only some apoptotic associate with membrane phospholipids. Membrane associ- stimuli. In addition, it is unclear whether release of cathepsins ation occurs in response to certain cellular stimuli, and this from lysosomes is controlled by pores or translocators, or is association may lower the intrinsic Ca++ requirements of these simply the consequence of damage to lysosomal membranes enzymes.67 A third factor influencing calpain activity may be during the apoptotic process. autolysis. Following the binding of Ca++, calpains undergo To date, relatively little is known about potential intersec- autolysis, with cleavages occurring near the amino terminii of tions of cathepsin- and caspase-mediated pathways, although the large and small subunits.68–73 Autolysis appears to increase it seems likely that these pathways will be integrated in some activity and lower the requirement for Ca++. Although it fashion. One report has shown that cathepsin G, which is remains controversial as to whether autolysis is absolutely abundantly expressed in neutrophils, can cleave and activate required for activation, Western blotting, looking for conver- procaspase-7 in vitro.53 Cathepsin G cleaves procaspase-7 sion of calpains to lower molecular weight forms, is a con- between the large and small subunits in the zymogen mol- venient and frequently used means of assessing calpain acti- ecule. Another report has demonstrated that cathepsin B can vation in cells. Finally, a fourth factor influencing calpain readily cleave caspase-1 and procaspase-11.52 In addition, activity is the presence of , a 110 kDa endogenous cathepsin B showed weak cleavage activity towards procas- protein inhibitor.74,75 Calpastatin is commonly expressed and pase-2, -6, -7 and -14. It was not determined, however, is highly specific for calpains; it is not known to inhibit any whether cathepsin B-mediated cleavage of procaspases other proteases. The expression and function of calpastatin resulted in the activation of these enzymes. Also, while may be regulated during apoptosis by other proteases, includ- cathepsin G and cathepsin B clearly can cleave certain pro- ing caspases (discussed below). caspases in vitro, it is not known whether cathepsin-mediated Calpains have been implicated in apoptosis based on two cleavage of caspases occurs in whole cells. Thus, it is difficult types of observations: (1) the activation of calpains during cell to establish whether caspase activation may be upstream or death and (2) the inhibition of apoptotic execution by various downstream of cathepsin activation during apoptosis. Perhaps calpain inhibitors. Calpain activation has been assessed using the use of highly selective inhibitors will help to address these several different assays including, Western blotting to detect issues. In this regard, however, a caveat should be raised. Pep- calpain autolysis, Western blotting to detect cleavage of tides based on caspase cleavage sequences, and derivatized known calpain substrate proteins, and in vitro and whole cell with fluoromethyl ketone or chloromethyl ketone have com- assays to detect cleavage of the fluorogenic calpain substrate monly been used as specific inhibitors of caspases in vitro and N-succinyl-leu-leu-val-tyr-7-amido-4-methylcourmarin (N-s- in whole cells. Strikingly, a recent study found that z-VAD- LLVY-AMC). Using these and a few less common assays, cal- FMK, z-DEVD-FMK, and Ac-YVAD-CMK potently inhibited pain activation has been detected in response to a variety of cathepsin B activity both in vitro and in cells.54 Therefore, the apoptotic stimuli, in a variety of different cell types. For impact of these inhibitors on biochemical events may be due example, in murine thymocytes treated with dexamethasone, to inhibition of either caspases or cathepsins, or both. calpain activity increases rapidly, peaking within 1 h of treat- ment.76 Also, in freshly isolated, apoptosis-prone, human neu- trophils, calpain enzymes are found to be constitutively Calpains and apoptosis active.77 Treatment of BL30A Burkitt’s lymphoma cells with radiation induces rapid calpain activation (15 min), as meas- Calpains comprise a family of cytoplasmic neutral cysteine ured by cleavage of the calpain substrate fodrin.78 By contrast, proteases.55–59 Both ubiquitously expressed and tissue-specific incubation of HL-60 cells with 9-amino-20(S)-camptothecin calpains have been identified.58,60,61 The most ubiquitous cal- causes slow autolysis/activation of calpain (8–10 h) compared pains are µ-calpain (or calpain I) and m-calpain (calpain II). with rapid activation of caspase-3 (2 h;79). Activation of cal- The µ- and m-calpains have been the focus of most studies pains has also been observed in nonhematopoietic cells. In involving calpains, and this review will focus exclusively on neuronally differentiated PC12 cells, induction of apoptosis by these two isoforms. It should be kept in mind, however, that ceramide analogs stimulates calpain-mediated cleavage of N- tissue-specific calpains may have essential in vivo roles in s-LLVY-AMC.80 Similar activation of N-s-LLVY-AMC cleavage apoptosis execution. In addition, calpain homologs have been activites are seen during apoptosis caused by reovirus infec- identified in organisms such as C. elegans, where genetic tion of cultured fibroblasts (81) or in vivo ischemia-reperfusion analyses may provide clues regarding the importance of injury in rat hepatocytes.82 these proteases.62 In addition to observations of calpain activation in experi- Both µ-calpain and m-calpain are composed of two sub- mental apoptosis, activation or overexpression of calpains has units, a large catalytic subunit of approximately 80 kDa, and also been seen in human diseases marked by excessive cell a smaller subunit of approximately 30 kDa.58,63 The 80 kDa death. For example, considerable activation of calpains is subunits specific for µ-calpain and m-calpain are encoded by seen in tissue from patients with Alzheimer’s disease distinct , while the 30 kDa subunit is shared between the when compared with normal brain tissue.83,84 Moreover, two isoforms. Both enzymes bind and require Ca++ for optimal Alzheimer’s also exhibit significantly reduced activity.55–57,59,64,65 Although µ- and m-calpains appear to expression of the calpain inhibitor calpastatin.84 In Parkinson’s have the same substrate specificity, they are functionally dis- disease, elevated expression of m-calpain is found in the tinquished on the basis of their Ca++ sensitivity. µ-Calpain mesencephalon region of the brain.85 Thus, it is possible that requires micromolar concentrations of Ca++ for optimal overzealous expression and/or activation of calpains may activity, while m-calpain requires millimolar concentrations. contribute to inappropriate cell loss in some important patho- The activation and activity of calpains is influenced by sev- logical conditions. eral factors. As mentioned, calpains must bind Ca++ in order While the activation of calpains during apoptosis suggests to be active, and sequences resembling EF-hand Ca++ binding an involvement of these enzymes in the execution process, it motifs are found in both the 80 and 30 kDa subunits.66 does not directly demonstrate an essential role. An under-

Leukemia Review DE Johnson 1698 standing of the importance of calpains in apoptotic execution amyloid peptide or staurosporin.98,99 Taken together, the stud- has come from studies using calpain inhibitors. A number of ies described above show that calpains can play important different calpain inhibitors have been developed. Unfortu- roles in the apoptotic execution of both hematopoietic and nately, many of the earlier inhibitors are somewhat nonspe- nonhematopoietic cell types. cific. Thus, experiments using these inhibitors must be inter- The ability of calpains to promote apoptosis in a variety of preted with caution; it is possible that actions ascribed to systems raises questions regarding the molecular calpains may actually be due to other cross-inhibited pro- mechanism(s) of calpain action. Undoubtedly, the cleavage of teases (eg the proteasome or cysteine proteases in the cathep- specific substrate proteins accounts for the proapoptotic sin family). The first generation of calpain inhibitors included activity of calpain, since the catalytic activity of the enzyme the active site inhibitors and E64 (E64d is the cell is required. A large variety of proteins have been shown to be permeable derivative of E64).86–89 These inhibitors also exhibit calpain substrates, including , alpha-actinin, , fila- 77,100 94,101 96 102 β 103 considerable activity against the proteasome and lysosomal min, fodrin, , FAK, integrin 3, PKC cysteine proteases. More recent active site inhibitors include α/β/γ,104 /-dependent IV,105 c- calpain inhibitor I (N-acetyl-leu-leu-norleucinal (aLLnL)), cal- mos,106 c-fos, c-jun,107,108 cyclin D1,109 p53,110 procaspase-3 pain inhibitor II (N-acetyl-leu-leu-methioninal (aLLM)), and and -9,96 and Bax.111 This group of substrate proteins is clearly calpeptin (benzyloxycarbonyl-leu-norleucinal).88–90 Although very diverse. However, it should be noted that several of these more specific than leupeptin and E64, these inhibitors also proteins are cytoskeletal proteins or proteins that can associate inhibit to some degree lysosomal cysteine proteases and the with cell membranes. This has led to speculation that calpains proteasome. An even more specific calpain inhibitor is the may be particularly important in destruction of cellular archi- compound PD150606 [3-(4-iodophenyl)-2-mercapto-(Z)-2- tecture during apoptosis. The impact of calpain-mediated propenoic acid], which interacts with Ca++ binding sites in the cleavage of the apoptosis regulatory proteins p53, Bax, and calpain enzymes.91 Finally, the most specific calpain inhibitor procaspase-3 and -9 is presently unclear. In the case of known to exist is the protein calpastatin, which directly binds caspase-3 and -9, calpain cleavage neither activates nor to calpains and potently inhibits their activities.74,75 To date, inactivates the caspase enzyme.96 Unfortunately, many of the however, relatively few studies have employed calpastatin to proteins identified as calpain substrates have only been shown investigate the role of calpains in apoptotic cell death. to be cleaved using in vitro experiments. Thus, it remains As perhaps may have been expected, numerous studies unclear how many of these proteins are directly cleaved by have shown that calpain inhibitors inhibit apoptosis in calpains in vivo. Also, it is currently unknown which sub- response to some, but not all, apoptotic stimuli. Studies by strates may be priority substrates, whose cleavage is critical Squier et al76,92 showed that dexamethasone-induced to the proapoptotic action of calpain. apoptosis of mouse thymocytes was inhibited by E64d and A number of studies have indicated cross-talk between cal- calpain inhibitor I. It was also found that calpain inhibitor I pain-mediated pathways and caspase-mediated pathways. In could inhibit thymocyte apoptosis caused by low-dose radi- some cases, caspase activation has been reported to be ation, A23187 ionophore, ionomycin, and forskolin; each of upstream of calpain activation, while in other cases the these forms of apoptosis requires new RNA synthesis in the opposite has been found. Wood and Newcomb79 determined thymocyte.92 By contrast, calpain inhibitor I could not prevent that calpain activation in camptothecin-treated HL-60 cells apoptosis that is not dependent on RNA synthesis, namely can be blocked by the pancaspase inhibitor z-VAD-FMK. By apoptosis caused by heat shock or valinomycin. In human contrast, the calpain inhibitor calpeptin failed to block acti- neutrophils, calpain inhibitor I and PD150606 were shown to vation of caspases. Interestingly, two groups have reported inhibit apoptosis caused by cycloheximide treatment, but not that activated caspases cleave and inactivate calpastatin apoptosis resulting from Fas stimulation.93 During spon- inhibitor.112,113 Caspase-mediated calpastatin cleavage was taneous neutrophil apoptosis, inhibition of calpains by calpep- seen in Jurkat and U937 cells following stimulation of Fas or tin alone, or the proteasome-specific inhibitor, lactacystin, TNF receptors, or following treatment with staurosporin. It alone had no effect on the rate of cell death.77 However, when remains undetermined, however, whether cleavage of calpas- used in combination, these inhibitors displayed a synergistic tatin inhibitor is entirely responsible for the calpain activation inhibitory effect, indicating synergy between calpains and the in these cells. An opposite relationship between calpains and proteasome in spontaneous neutrophil apoptosis. Related caspases has been reported during radiation-induced studies have revealed that antisense-mediated downregulation apoptosis. Waterhouse et al78 observed that following treat- of calpastatin inhibitor protein significantly accelerates neu- ment of BL30A lymphoma cells with radiation, calpain was trophil apoptosis.93 Studies with HL-60 cells have shown that activated early, and caspases were activated significantly later. calpain inhibition by calpeptin fails to block DNA fragmen- Moreover, inhibition of calpain activity abrogated down- tation and loss of viability caused by treatment with campto- stream activation of caspase-3. In yet another study, calpain thecin.79 However, calpeptin was able to block TNF-induced activation was found to occur independently of caspase acti- U937 cell apoptosis, while calpain inhibitor I delayed U937 vation during activation of platelets.96 Given these different apoptosis resulting from calphostin treatment.94,95 In human findings, it seems likely that the relationship between caspases platelets, calpeptin blocked A23187-induced caspase frag- and calpains may vary depending on the cell type and apop- mentation, cleavage of apoptosis substrate proteins, and totic stimulus. microvesiculation.96 Studies with nonhematopoietic cells have shown that calpain inhibitor I and PD150606 blocked reovirus-induced apoptosis in murine fibroblasts, and calpain Granzyme B and apoptosis inhibitors I and II blocked TGF-β-induced DNA fragmentation and loss of viability in rat hepatocytes.81,97 In addition, calpain Cytotoxic T lymphocytes (CTLs) and natural killer (NK) cells inhibitor I blocked apoptosis in chick ciliary neurons deprived are important for the host defense against viruses, parasitic of CNTF, and the calpain inhibitor MDL 28,170 inhibited the agents, and transformed cells.114,115 CTLs and NK cells induce death of rat hippocampal pyramidal neurons treated with β- apoptosis in target cells using at least two distinct mech-

Leukemia Review DE Johnson 1699 anisms. One mechanism involves stimulation of cell surface DEVD- or VAD-containing (100 µM DEVD-CHO or death receptors (such as Fas) on the target cells by death VAD peptides inhibit caspases, but not granzyme B).139,151,153–155 116,117 ligands expressed on the surface of the effector cell. This Two additional caspase substrate proteins, DNA-PKcs and leads to activation of caspase cascades in the target cell. NuMA, are also cleaved in granzyme B/perforin-treated cells, Another mechanism, termed ‘granule exocytosis’, involves the but cleavage of these proteins is insensitive to DEVD or VAD 155 vectoral transfer of the contents of effector cell cytoplasmic peptide inhibitors. Moreover, the sizes of the DNA-PKcs and granules into the target cell.118–120 Key components of these NuMA proteolytic fragments generated by granzyme B differ granules are perforin and the granzyme family of serine pro- from those resulting from caspase cleavage. This indicates that teases. during granzyme B-mediated apoptosis, important cellular Perforin is a 70 kDa protein that binds to membrane phos- substrates are cleaved in a caspase-independent fashion. The phorylcholine groups in a calcium-dependent manner.121–123 importance of these caspase-independent cleavage events Following binding, perforin inserts into the membrane and oli- remains to be determined. However, the fact that granzyme gomerizes, forming pores. This permeabilization of the mem- B/perforin-mediated DNA fragmentation and apoptotic death brane may facilitate the entry of other molecules, including is significantly delayed by 100 µm DEVD/VAD,139,153,154 granzymes, into the target cell. underscores the necessity for caspase activation during this Within the granules of CTLs and NK cells, granzymes A and form of apoptosis. B are particularly abundant.124 Granzyme B (also called frag- mentin or protease (CCP)) shares with casp- ases the unique characteristic of cleaving substrate proteins Granzyme A and apoptosis after aspartate residues.125–129 An important role for granzyme B in the induction of target cell apoptosis has been demon- Granzyme A is the most abundant protease found in the gran- strated using gene knockout mice. CTLs and NK cells derived ules of CTL cells. The mature granzyme A enzyme is a disul- from granzyme B−/− mice exhibit greatly reduced capacity to phide cross-linked homodimer of 50 kDa that cleaves sub- induce apoptotic DNA fragmentation in target cells.119,130 strate proteins following lysine or arginine residues.127,156,157 Earlier complementary studies showed that purified granzyme Although granzyme A is capable of inducing apoptosis after B alone did not promote apoptosis when added to target cells. loading into target cells, the mechanism of action of this pro- However, cotreatment with purified granzyme B and perforin tease differs significantly from that of granzyme B. In addition, proteins induced marked DNA fragmentation and apoptotic based on the experimental systems that have been employed features in four lymphoma target cell lines.129 Based on the thus far, it appears that the role of granzyme A in CTL-induced ability of perforin to form membrane pores, it has been the apoptosis is far more subtle than that of granzyme B. Mice general consensus that granzyme B gains entry into target cells which are deficient in granzyme A expression (granzyme A−/− through perforin pores. This mode of entry, however, is cur- mice), exhibit relatively normal CTL-mediated cytotoxicity.158 rently being called into question. Several studies have shown The only reported defect for granzyme A−/− mice is an inability that granzyme B is internalized by target cells in the absence to clear the mouse pox virus Ectromelia.159 By contrast, CTLs of added perforin.131–135 The internalized granzyme B has from granzyme B−/− mice are capable of inducing target cell been reported to reside in the cytoplasm,132,133 or in a novel death only after prolonged coincubation.130 Thus, granzyme vesicular compartment.134 The triggering of apoptosis in cells B is critically important for rapid CTL killing. The possibility that have internalized granzyme B requires further addition of that granzyme A does have some role in CTL-mediated killing, perforin to the cells.131–135 It may be that perforin is required has been suggested by recent experiments using mice that are for the release of granzyme B from internal vesicles in the deficient in both granzyme A and granzyme B. CTLs from target cell. Other studies have indicated that perforin facili- granzyme A−/−/granzyme B−/− mice are unable to induce target tates translocation of granzyme B to the nucleus, and that cell DNA fragmentation, even after prolonged coincu- nuclear localization is critical to the ability of granzyme B to bation.160 This indicates that granzyme A activity accounts for cause apoptosis.132–137 the ability of granzyme B−/− CTLs to induce target cell While the importance of granzyme B subcellular localiz- apoptosis after prolonged exposure. Therefore, granzyme A ation remains controversial, it is quite clear that granzyme B may allow CTLs to kill target cells under conditions where has the ability to impact the caspase pathway of apoptosis. granzyme B activity is inhibited (eg target cells that express Studies done in vitro, have shown that granzyme B is capable granzyme B inhibitors). of cleaving procaspase-3, -6, -7, -8, -9 and -10.138–153 In the Studies using recombinant proteins have shown that coin- case of procaspases-3, -7 and -9, granzyme B-mediated pro- cubation of granzyme A and perforin with target cells leads cessing has been shown to generate active caspase to rapid (within 2 h) accumulation of DNA single-strand enzymes.138,141,146,149 More importantly, studies with whole breaks.161,162 This contrasts with the rapid degradation of cells have shown that caspases are activated in target cells DNA to oligonucleosomal-length fragments seen in cells following coincubation with granzyme B and per- treated with granzyme B and perforin. Granzyme A/perforin forin.139,153,154 It remains to be determined, however, which treatment also leads to nuclear condensation.162 The DNA sin- caspases are the preferred in vivo substrates for granzyme B. gle-strand breakage and nuclear condensation that occurs in In any event, it is reasonable to propose that granzyme B may response to granzyme A is insensitive to caspase inhibitors, promote apoptosis simply by cleaving and activating indicating that these actions of granzyme A are caspase-inde- endogenous caspases in the target cell. pendent.162 Consistent with this, granzyme A/perforin treat- In cells undergoing granzyme B/perforin-mediated ment does not result in processing/activation of procaspase-3 apoptosis, cleavage of the caspase substrate proteins PARP, or cleavage of the caspase substrate proteins PARP, B, lamin B, and U1–70 kDa is also observed.151,153–155 These or rho-GTPase.162 By contrast, granzyme B-induced DNA cleavage events appear to be due to caspases activated by fragmentation is strictly dependent on the activation of casp- granzyme B, and not the result of direct granzyme B cleavage, ases. Both granzyme A and granzyme B (in conjunction with since cleavage of all three proteins is inhibited by 100 µM perforin) also induce target cell cytolysis, and in both cases

Leukemia Review DE Johnson 1700 this is a caspase-independent event. Taken together, current 21 Tandon AK, Clark GM, Chamness GC, Chirgwin JM, McGuire WL. evidence indicates that granzyme B is the primary CTL Cathepsin D and prognosis in breast cancer. New Engl J Med mediator of target cell DNA fragmentation and apoptotic 1990; 322: 297–302. 22 Kute TE, Shao ZM, Sugg NK, Long RT, Russell GB, Case D. death, and that the apoptotic effects of this protease are Cathepsin D as a prognostic indicator of node-negative breast mediated primarily through the activation of caspases. Gran- cancer patients using both immunoassays and enzymatic assays. zyme A, on the other hand, may be more of a default or speci- Cancer Res 1992; 52: 198–203. alized mediator of target cell apoptosis, with the pathways 23 Garcia M, Platet N, Liaudet E, Laurent V, Derocq D, Brouillet J- initiated by granzyme A being distinctly different from those P, Rochefort H. Biological and clinical significance of cathepsin initiated by granzyme B. D in breast cancer metastasis. Stem Cells 1996; 14: 642–650. 24 English HF, Kyprianou N, Isaacs JT. Relationship between DNA fragmentation and apoptosis in in the rat prostate following castration. Prostate 1989; 15: 233–250. References 25 Walker NI, Bennett RE, Kerr JF. Cell death by apoptosis during involution of the lactating breast in mice and rats. Am J Anat 1989; 1 Yuan J, Shaham S, Ledoux S, Ellis HM, Horvitz HR. The C. elegans 185: 19–32. cell death gene ced-3 encodes a protein similar to mammalian β 26 Guenette RS, Mooibroek M, Wong K, Wong P, Tenniswood M. interleukin-1 -converting enzyme. Cell 1993; 75: 641–652. Cathepsin B, a cysteine protease implicated in metastatic pro- 2 Cerretti DP, Kozlosky CJ, Mosley B, Nelson N, Van Ness K, Green- gression, is also expressed during regression of the rat prostate and street TA, March CJ, Kronheim SR, Druck T, Cannizzaro LA, β mammary glands. Eur J Biochem 1994; 226: 311–321. Huebner K, Black RA. Molecular cloning of the interleukin-1 27 Sensibar JA, Liu XX, Patai B, Alger B, Lee C. Characterization of converting enzyme. Science 1992; 256: 97–100. castration-induced cell death in the rat prostate by immunohisto- 3 Thornberry NA, Bull HG, Calaycay JR, Chapman KT, Howard AD, chemical localization of cathepsin D. Prostate 1990; 16: 263–276. Kostura MJ, Miller DK, Mokineaux SM, Weidner JR, Aunins J, Ellis- 28 Roberts LR, Kurosawa H, Bronk SF, Fesmier PJ, Agellon LB, Leung ton KO, Ayala JM, Casano FJ, Chin J, Ding GJ-F, Egger LA, Gaffney W-Y, Mao F, Gores GJ. Cathepsin B contributes to bile salt- EP, Limjuco G, Palyha OC, Raju SM, Rolando AM, Salley JP, induced apoptosis of rat hepatocytes. Gastroenterology 1997; Yamin T-T, Lee TD, Shively JE, MacCross M, Mumford RA, 113: 1714–1726. Schmidt JA, Tocci MJ. A novel heterodimeric cysteine protease is 29 Roberts LR, Adjei PN, Gores GJ. Cathepsins as effector proteases required for interleukin-1β processing in monocytes. Nature 1992; in hepatocyte apoptosis. Cell Biochem Biophys 1999; 30: 71–88. 356: 768–774. 30 Nishimura Y, Kawabata T, Kato K. Identification of latent cathep- 4 Alnemri ES, Livingston DJ, Nicholson DW, Salvesen G, Thornberry sins B and L in microsomal lumen: characterization of enzymatic NA, Wong WW, Yuan J. Human ICE/CED-3 protease nomencla- activation and proteolytic processing in vitro. Arch Biochem ture. Cell 1996; 87: 171. Biophys 1988; 261: 64–71. 5 Thornberry NA, Lazebnik Y. Caspases: enemies within. Science 31 Rowan AD, Mason P, Mach L, Mort JS. Rat procathepsin B: Proteo- 1998; 281: 1312–1316. lytic processing to the mature form in vitro. J Biol Chem 1992; 6 Orlowski RZ. The role of the ubiquitin-proteasome pathway in 267: 15993–15999. apoptosis. Cell Death Differ 1999; 6: 303–313. 7 Schwartz MK. Tissue cathepsins as tumor markers. Clin Chem Acta 32 Shibata M, Kanamori S, Isahara K, Ohsawa Y, Konishi A, Kame- 1995; 237: 67–78. taka S, Watanabe T, Ebisu S, Ishido K, Kominami E, Uchiyama Y. 8 Chapman HA, Riese RJ, Shi GP. Emerging roles for cysteine pro- Participation of cathepsins B and D in apoptosis of PC12 cells teases in human biology. Ann Rev Physiol 1997; 59: 63–88. following serum deprivation. Biochem Biophys Res Commun 9 Westley B, Rochefort H. A secreted glycoprotein induced by estro- 1998; 251: 199–203. gen in human breast cancer cell lines. Cell 1980; 20: 353–362. 33 Cataldo AM, Barnett JL, Berman SA, Li J, Quarless S, Bursztajn S, 10 Capony F, Rougeot C, Montcourrier P, Cavailles V, Salazar G, Lippa C, Nixon RA. and cellular content of Rochefort H. Increased secretion, altered processing and glycosyl- cathepsin D in Alzheimer’s disease brain: evidence for early up- ation of procathepsin D in mammary cancer cells. Cancer Res regulation of the endosomal-lysosomal system. 1995; 14: 1989; 49: 3904–3909. 671–680. 11 Erickson AH. Biosynthesis of lysosomal endopeptidases. J Cell 34 Isahara K, Ohsawa Y, Kanamori S, Shibata M, Waguri S, Sato N, Biochem 1989; 40: 31–41. Gotow T, Watanabe T, Momoi T, Urase K, Kominami E, Uchiyama 12 Fujita H, Tanaka Y, Noguchi Y, Kono A, Himeno M, Kato K. Iso- Y. Regulation of a novel pathway for cell death by lysosomal lation and sequencing of a cDNA clone encoding rat liver lysoso- aspartic and cysteine proteinases. Neuroscience 1999; 91: 233– mal cathepsin D and the structure of three forms of mature 249. enzymes. Biochem Biophys Res Commun 1991; 179: 190–196. 35 Lotem J, Sachs L. Control of apoptosis in hematopoiesis and leuke- 13 Godbold GD, Ahn K, Yeyeodu S, Lee LF, Ting JP, Erickson AH. mia by cytokines, tumor suppressor and oncogenes. Leukemia Biosynthesis and intracellular targeting of the lysosomal aspartic 1996; 10: 925–931. proteinase cathepsin D. Adv Exp Med Biol 1998; 436: 153–162. 36 Drexler HG, Zaborski M, Quentmeier H. Cytokine response pro- 14 Kornblau SM. The role of apoptosis in the pathogenesis, prognosis, files of human myeloid factor-dependent leukemia cell lines. Leu- and therapy of hematologic malignancies. Leukemia 1998; 12 kemia 1997; 11: 701–708. (Suppl 1): 41–46. 37 Antoku K, Liu Z, Johnson DE. IL-3 withdrawal activates a CrmA- 15 Mort JS, Recklies AD. Interrelationship of active and latent insensitive poly(ADP-ribose) polymerase cleavage enzyme in fac- secreted human cathepsin B precursors. Biochem J 1986; 233: tor-dependent myeloid progenitor cells. Leukemia 1998; 12: 57–63. 682–689. 16 Sloane BF, Honn KV. Cysteine proteinases and metastasis. Cancer 38 Blalock WL, Weinstein-Oppenheimer C, Chang F, Hoyle PE, Metast Rev 1984; 3: 249–263. Wang XY, Algate PA, Franklin RA, Oberhaus SM, Steelman LS, 17 Briozzo P, Morisset M, Capony F, Rougeot C, Rochefort H. In vitro McCubrey JA. Signal transduction, regulatory, and anti- degradation of extracellular matrix with Mr 52,000 cathepsin D apoptotic pathways regulated by IL-3 in hematopoietic cells: poss- secreted by breast cancer cells. Cancer Res 1988; 48: 3688–3692. ible sites for intervention with anti-neoplastic drugs. Leukemia 18 Leto G, Gebbia N, Rausa L, Tumminello FM. Cathepsin D in the 1999; 13: 1109–1166. malignant progression of neoplastic disease. Anticancer Res 1992; 39 Deiss LP, Galinka H, Berissi H, Cohen O, Kimchi A. Cathepsin D 12: 235–240. protease mediates programmed cell death induced by - 19 Mignatti P, Rifkin DB. Biology and biochemistry of proteinases in γ, Fas/APO-1 and TNF-α. EMBO J 1996; 15: 3861–3870. tumor invasion. Physiol Rev 1993; 73: 161–195. 40 Wu GS, Saftig P, Peters C, El-Deiry WS. Potential role for cathep- 20 Thorpe SM, Rochefort H, Garcia M, Freiss G, Christensen IJ, Khalaf sin D in p53-dependent tumor suppression and chemosensitivity. S, Paolucci F, Pau B, Rasmussen BB, Rose C. Association between Oncogene 1998; 16: 2177–2183. high concentration of 52-kDa cathepsin D and poor prognosis in 41 Luo X, Budihardjo I, Zou H, Slaughter C, Wang X. Bid, a Bcl2 primary human breast cancer. Cancer Res 1989; 49: 6008–6014. interacting protein, mediates release from mito-

Leukemia Review DE Johnson 1701 chondria in response to activation of cell surface death receptors. of calpains I and II from various sources as demonstrated by Cell 1998; 94: 481–490. immunological cross-reactivity. J Biochem 1983; 94: 2055–2061. 42 Li H, Zhu H, Xu C-J, Yuan J. Cleavage of BID by 64 Murachi T. Intracellular regulatory system involving calpain and mediates the mitochondrial damage in Fas pathway of apoptosis. calpastatin. Biochem Int 1989; 18: 263–294. Cell 1998; 94: 491–501. 65 Croall DE, DeMartino GN. Calcium-activated neutral protease 43 Friesen C, Fulda S, Debatin KM. Cytotoxic drugs and the CD95 (calpain) system: Structure, function, and regulation. Physiol Rev pathway. Leukemia 1999; 13: 1854–1858. 1991; 71: 813–847. 44 Adams JM, Cory S. The Bcl-2 : arbiters of cell sur- 66 Blanchard H, Grochulski P, Li Y, Arthur JS, Davies PL, Elce JS, vival. Science 1998; 281: 1322–1326. Cygler M. Structure of a calpain Ca(2+)-binding domain reveals a 45 Liu X, Zou H, Slaughter C, Wang X. DFF, a heterodimeric protein novel EF-hand and Ca(2+)-induced conformational changes. Nat- that functions downstream of caspase-3 to trigger DNA fragmen- ure Struct Biol 1997; 4: 532–538. tation during apoptosis. Cell 1997; 89: 175–184. 67 Molinari M, Anagli J, Carafoli E. Ca(2+)-activated neutral protease 46 Enari M, Sakahira H, Yokoyama H, Okawa K, Iwamatsu A, Nagata is active in the erythrocyte membrane in its nonautolyzed 80-kDa S. A caspase-activated DNase that degrades DNA during form. J Biol Chem 1994; 269: 27992–27995. apoptosis, and its inhibitor ICAD. Nature 1998; 391: 43–50. 68 Suzuki K, Tsuji S, Ishiura S, Kimura Y, Kubota S, Imahori K. Autoly- 47 Rowan S, Fisher DE. Mechanisms of apoptotic cell death. Leuke- sis of calcium-activated neutral protease of chicken skeletal mus- mia 1997; 11: 457–465. cle. J Biochem 1981; 90: 1787–1793. 48 Wolf BB, Green DR. Suicidal tendencies: apoptotic cell death by 69 Mellgren RL, Repetti A, Muck TC, Easly J. Rabbit caspase family proteinases. J Biol Chem 1999; 274: 20049–20052. calcium-dependent protease requiring millimolar CA2+. Purifi- 49 Brunk UT, Zhang H, Roberg K, Ollinger K. Lethal hydrogen per- cation, subunit structure, and Ca2+-dependent autoproteolysis. J oxide toxicity involves lysosomal iron-catalyzed reactions with Biol Chem 1982; 257: 7203–7209. membrane damage. Redox Rep 1995; 1: 267–277. 70 DeMartino GN, Huff CA, Croall DE. Autoproteolysis of the small 50 Li W, Yuan XM, Olsson AG, Brunk UT. Uptake of oxidized LDL subunit of calcium-dependent protease II activates and regulates by macrophages results in partial lysosomal enzyme inactivation protease activity. J Biol Chem 1986; 261: 12047–12052. and relocation. Arterioscler Thromb Vasc Biol 1998; 18: 177–184. 71 Imajoh S, Kawasaki H, Suzuki K. Limited autolysis of calcium- 51 Roberg K, Ollinger K. Oxidative stress causes relocation of the activated neutral protease (CANP): reduction of the Ca2+-require- lysosomal enzyme cathepsin D with ensuing apoptosis in neonatal ment is due to the NH2-terminal processing of the large subunit. rat cardiomyocytes. Am J Pathol 1998; 152: 1151–1156. J Biochem 1986; 100: 633–642. 52 Vancompernolle K, Van Herreweghe F, Pynaert G, Van de Craen 72 Inomata M, Kasai Y, Nakamura M, Kawashima S. Activation M, De Vos K, Totty N, Sterling A, Fiers W, Vandenabeele P, mechanism of calcium-activated neutral protease. Evidence for Grooten J. Atractyloside-induced release of cathepsin B, a pro- the existence of intramolecular and intermolecular autolyses. J tease with caspase-processing activity. FEBS Lett 1998; 438: Biol Chem 1988; 263: 19783–19787. 150–158. 73 Elce JS, Hegadorn C, Arthur JSC. Autolysis, Ca2+ requirement, and 53 Zhou Q, Salvesen GS. Activation of pro-caspase-7 by serine pro- heterodimer stability in m-calpain. J Biol Chem 1997; 272: teases includes a non-canonical specificity. Biochem J 1997; 324: 11268–11275. 361–364. 74 Waxman L, Krebs EG. Identification of two protease inhibitors 54 Schotte P, Declercq W, Van Huffel S, Vandenabeele P, Beyaert from bovine cardiac muscle. J Biol Chem 1978; 253: 5888–5891. R. Non-specific effects of methyl ketone peptide inhibitors of casp- 75 Emori Y, Kawasaki H, Imajoh S, Imahori K, Suzuki K. Endogenous ases. FEBS Lett 1999; 442: 117–121. inhibitor for calcium-dependent cysteine protease contains four 55 Guroff G. A neutral, calcium-activated proteinase from the soluble internal repeats that could be responsible for its multiple reactive fraction of rat brain. J Biol Chem 1964; 239: 149–155. sites. Proc Natl Acad Sci USA 1987; 84: 3590–3594. 56 Mellgren RL. Canine cardiac calcium-dependent proteases: resol- 76 Squier MKT, Miller ACK, Malkinson AM, Cohen JJ. Calpain acti- ution of two forms with different requirements for calcium. FEBS vation in apoptosis. J Cell Physiol 1994; 159: 229–237. Lett 1980; 109: 129–133. 77 Knepper-Nicolai B, Savill J, Brown SB. Constitutive apoptosis in 57 Murachi T, Tanaka K, Hatanaka M, Murakami T. Intracellular human neutrophils requires synergy between calpains and the Ca2+-dependent protease (calpain) and its high-molecular-weight proteosome downstream of caspases. J Biol Chem 1998; 273: endogenous inhibitor (calpastatin). Adv Enzyme Reg 1981; 19: 30530–30536. 407–424. 78 Waterhouse NJ, Finucane DM, Green DR, Elce JS, Kumar S, 58 Sorimachi H, Saido TC, Suzuki K. New era of calpain research: Alnemri ES, Litwack G, Khanna KK, Lavin MF, Watters DJ. Calpain discovery of tissue-specific calpains. FEBS Lett 1994; 341: 1–5. activation is upstream of caspases in radiation-induced apoptosis. 59 Carfoli E, Molinari M. Calpain: a protease in search of a function? Cell Death Differ 1998; 5: 1051–1061. Biochem Biophys Res Commun 1998; 247: 193–203. 79 Wood DE, Newcomb EW. Caspase-dependent activation of cal- 60 Sorimachi H, Ishiura S, Suzuki K. A novel tissue-specific calpain pain during drug-induced apoptosis. J Biol Chem 1999; 274: species expressed predominantly in the stomach comprises two 8309–8315. products with and without Ca(2+)-binding 80 Xie H, Johnson GV. Ceramide selectively decreases tau levels in domain. J Biol Chem 1993; 268: 19476–19482. differentiated PC12 cells through modulation of calpain I. J Neuro- 61 Sorimachi H, Toyama-Sorimachi N, Saido TC, Kawasaki H, Sugita chem 1997; 69: 1020–1030. H, Miyasaka M, Arahata K, Ishiura S, Suzuki K. Muscle-specific 81 Debiasi RL, Squier MKT, Pike B, Wynes M, Dermody TS, Cohen calpain, p94, is degraded by autolysis immediately after trans- JJ, Tyler KL. Reovirus-induced apoptosis is preceded by increased lation, resulting in disappearance from muscle. J Biol Chem 1993; cellular calpain activity and is blocked by calpain inhibitors. J 268: 10593–10605. Virol 1999; 73: 695–701. 62 Wilson R, Ainscough R, Anderson K, Baynes C, Berks M, Bonfield 82 Kohli V, Madden JF, Bentley RC, Clavien P-A. Calpain mediates J, Burton J, Connell M, Copsey T, Cooper J, Coulson A, Craxton ishemic injury of the liver through modulation of apoptosis and M, Dear S, Du Z, Durbin R, Favello A, Fraser A, Fulton L, Gardner . Gastroenterol 1999; 116: 168–178. A, Green P, Hawkins T, Hillier L, Jler M, Johnston L, Jones M, 83 Saito K, Elce JS, Hamos JE, Nixon RA. Widespread activation of Kershaw J, Kirsten J, Laisster N, Latreille P, Lightning J, Lloyd C, calcium-activated neutral proteinase (calpain) in the brain in Alzh- Mortimore B, O’Callaghan M, Parsons J, Percy C, Rifken L, Roopra eimer disease: a potential molecular basis for neuronal degener- A, Saunders D, Shownkeen R, Sims M, Smaldon N, Smith A, Smith ation. Proc Natl Acad Sci USA 1993; 90: 2628–2632. M, Sonnhammer E, Staden R, Sulston J, Thierry-Mieg J, Thomas 84 Nixon RA, Saito K-I, Grynspan F, Griffin WR, Katayama S, Honda K, Vaudin M, Vaughan K, Waterston R, Watson A, Weinstock L, T, Mohan PS, Shea TB, Beermann M. Calcium-activated neutral Wilkinson-Sproat J, Wohldman P. 2.2 Mb of contiguous nucleo- proteinase (calpain) system in aging and Alzheimer’s disease. Ann tide sequence from III of C. elegans. Nature 1994; NY Acad Sci 1994; 747: 77–91. 368: 32–38. 85 Mouatt-Prigent A, Karlsson JO, Agid Y, Hirsch EC. Increased M- 63 Sasaki T, Yoshimura N, Kikuchi T, Hatanaka M, Kitahara A, Sakih- calpain expression in the mesencephalon of patients with - ama T, Murachi T. Similarity and dissimilarity in subunit structures son’s disease but not in other neurodegenerative disorders involv-

Leukemia Review DE Johnson 1702 ing the mesencephalon: a role in nerve cell death? Neurosci 1996; of the c-mos proto-oncogene product by calpain on 73: 979–987. fertilization of Xenopus eggs. Nature 1989; 342: 505–511. 86 Aoyagi T, Takeuchi T, Matsuzaki A, Kawamura K, Kondo S. Leu- 107 Hirai S, Kawasaki H, Yaniv M, Suzuki K. Degradation of tran- peptins, new protease inhibitors from Actinomycetes. J Antibiotics scription factors, c-Jun and c-Fos, by calpain. FEBS Lett 1991; 1969; 22: 283–286. 287: 57–61. 87 Barrett AJ, Kembhavi AA, Hanada K. E-64 [L-trans-epoxysuccinyl- 108 Watt F, Molloy PL. Specific cleavage of transcription factors by leucyl-amido(4-guanidino)butane] and related epoxides as inhibi- the thiol protease, m-calpain. Nucleic Acids Res 1993; 21: tors of cysteine proteinases. Acta Biol Med Germ 1981; 40: 5092–5100. 1513–1517. 109 Choi YH, Lee SJ, Nguyen P, Jang JS, Lee J, Wu ML, Takano E, 88 Wang KK. Developing selective inhibitors of calpain. Trends Maki M, Henkart PA, Trepel JB. Regulation of cyclin D1 by cal- Pharm Sci 1990; 11: 139–142. pain protease. J Biol Chem 1997; 272: 28479–28484. 89 Mehdi S. Cell-penetrating inhibitors of calpain. Trends Biochem 110 Kubbutat MHG, Vousden KH. Proteolytic cleavage of human Sci 1991; 16: 150–153. p53 by calpain: a potential regulator of protein stability. Mol Cell 90 Tsujinaka T, Kajiwara Y, Kambayashi J, Sakon M, Higuchi N, Biol 1997; 17: 460–468. Tanaka T, Mori T. Synthesis of a new cell penetrating inhibitor 111 Wood DE, Thomas A, Devi LA, Berman Y, Beavis RC, Reed JC, (calpeptin). Biochem Biophys Res Commun 1988; 153: 1201– Newcomb EW. Bax cleavage is mediated by calpain during drug- 1208. induced apoptosis. Oncogene 1998; 17: 1069–1078. 91 Wang KK, Nath R, Posner A, Raser KJ, Buroker-Kilgore M, Haji- 112 Porn-Ares MI, Samali A, Orrenius S. Cleavage of the calpain mohammadreza I, Probert W, Marcoux FW, Ye Q, Takano E, inhibitor, calpastatin, during apoptosis. Cell Death Differ 1998; Hatanaka M, Maki M, Caner H, Collins JL, Fergus A, Lee KS, 5: 1028–1033. Lunney EA, Hays SJ, Yuen P. An alpha-mercaptoacrylic acid 113 Wang KKW, Posmantur R, Nadimpalli R, Nath R, Mohan P, derivative is a selective non-peptide cell-permeable calpain Nixon RA, Talanian RV, Keegan M, Herzog L, Allen H. Caspase- inhibitor and is neuroprotective. Proc Natl Acad Sci USA 1996; mediated fragmentation of calpain inhibitor protein calpastatin 93: 6687–6692. during apoptosis. Arch Biochem Biophys 1998; 356: 187–196. 92 Squier MKT, Cohen JJ. Calpain, an upstream regulator of thymo- 114 Berke G. The CTL’s kiss of death. Cell 1995; 81: 9–12. cyte apoptosis. J Immunol 1997; 158: 3690–3697. 115 Kagi D, Lederman B, Burki K, Zinkernagel RM, Hengartner H. 93 Squier MKT, Sehnert AJ, Sellins KS, Malkinson AM, Takano E, Molecular mechanisms of lymphocyte-mediated cytotoxicity and Cohen JJ. Calpain and calpastatin regulate neutrophil apoptosis. their role in immunological protection and pathogenesis in vivo. J Cell Physiol 1999; 178: 311–319. Ann Rev Immunol 1996; 14: 207–232. 94 Vanags DM, Porn-Ares I, Coppola S, Burgess DH, Orrenius S. 116 Nagata S, Golstein P. The Fas death factor. Science 1995; 267: Protease involvement in fodrin cleavage and phosphatidylserine 1449–1456. exposure in apoptosis. J Biol Chem 1996; 271: 31075–31085. 117 Ashkenazi A, Dixit VM. Death receptors: signaling and modu- 95 Spinedi A, Oliverio S, Di Sano F, Piacentini M. Calpain involve- lation. Science 1998; 281: 1305–1308. ment in calphostin C-induced apoptosis. Biochem Pharm 1998; 118 Doherty PC. Cell-mediated cytotoxicity. Cell 1993; 75: 607–612. 56: 1489–1492. 119 Shresta S, MacIvor DM, Heusel JW, Russell JH, Ley TJ. Natural 96 Wolf BB, Goldstein JC, Stennicke HR, Beere H, Amarante- killer and lymphokine-activated killer cells require granzyme B Mendes GP, Salvesen GS, Green DR. Calpain functions in a cas- for the rapid induction of apoptosis in susceptible target cells. pase-independent manner to promote apoptosis-like events dur- Proc Natl Acad Sci USA 1995; 92: 5679–5683. ing platelet activation. Blood 1999; 94: 1683–1692. 120 Shresta S, Heusel JW, Macivor DM, Wesselschmidt RL, Russell 97 Gressner AM, Lahme B, Roth S. Attenuation of TGF-beta-induced JH, Ley TJ. Granzyme B plays a critical role in cytotoxic lympho- cyte-induced apoptosis. Immunol Rev 1995; 146: 211–221. apoptosis in primary cultures of hepatocytes by calpain inhibi- 121 Masson D, Tschopp J. Isolation of a lytic pore-forming protein tors. Biochem Biophys Res Commun 1997; 231: 457–462. (perforin) from cytolytic T lymphocytes. J Biol Chem 1985; 260: 98 Villa PG, Henzel WJ, Sensenbrenner M, Henderson CE, 9069–9072. Pettmann B. Calpain inhibitors, but not caspase inhibitors, pre- 122 Young JD-E, Hengartner H, Podack ER, Cohn ZA. Purification vent actin proteolysis and DNA fragmentation during apoptosis. and characterization of a cytolytic pore-forming protein from J Cell Sci 1998; 111: 713–722. granules of cloned lymphocytes with natural killer activity. Cell 99 Jordan J, Galindo MF, Miller RJ. Role of calpain- and interleukin- 1986; 44: 849–859. 1β converting enzyme-like proteases in the β-amyloid-induced 123 Tschopp J, Schafer S, Masson D, Peitsch MC, Heusser C. Phos- death of rat hippocampal neurons in culture. J Neurochem 1997; phorylcholine acts as a calcium dependent molecule for 68: 1612–1621. lymphocyte perforin. Nature 1989; 337: 272–274. 100 Yuan Y, Dopheide SM, Ivanidis C, Salem HH, Jackson SP. Cal- 124 Smyth MJ, O’Connor MD, Trapani JA. Granzymes: A variety of pain regulation of cytoskeletal signaling complexes in von Wille- serine protease specificities encoded by genetically distinct brand factor-stimulated platelets. Distinct roles for glycoprotein subfamilies. J Leuk Biol 1996; 60: 555–562. Ib-V-IX and glycoprotein IIb-IIIa (integrin alphaIIbbeta3) in von 125 Zunino SJ, Bleackley RC, Martinez J, Hudig D. RNKP-1, a novel Willebrand factor-induced signal transduction. J Biol Chem natural killer cell-associated serine protease gene cloned from 1997; 272: 21847–21854. RNK-16 cytotoxic lymphocytes. J Immunol 1990; 144: 2001– 101 Martin SJ, O’Brien GA, Nishioka WK, McGahon AJ, Mahboubi 2009. A, Saido TC, Green DR. Proteolysis of fodrin (non-erythroid 126 Lobe CG, Havele C, Bleackley RC. Cloning of two genes that ) during apoptosis. J Biol Chem 1995; 270: 6425–6428. are specifically expressed in activated cytotoxic T lymphocytes. 102 Cooray P, Yuan Y, Schoenwaelder SM, Mitchell CA, Salem HH, Proc Natl Acad Sci USA 1986; 83: 1448–1452. Jackson SP. kinase (pp125FAK) cleavage and 127 Odake S, Kam CM, Narasimhan L, Poe M, Blake JT, Krahenbuhl regulation by calpain. Biochem J 1996; 318: 41–47. O, Tschopp J, Powers JC. Human and murine cytotoxic T lym- 103 Meredith J Jr, Mu Z, Saido T, Du X. Cleavage of the cytoplasmic phocyte serine proteases: subsite mapping with peptide thioester β domain of integrin 3 subunit during endothelial cell apoptosis. substrates and inhibition of enzyme activity and cytolysis by isoc- J Biol Chem 1998; 273: 19525–19531. oumarins. Biochem 1991; 30: 2217–2227. 104 Kishimoto A, Mikawa K, Hashimotos K, Yasuda I, Tanaka S, Tom- 128 Poe M, Blake JT, Boulton DA, Gammon M, Sigal NH, Wu JK, inaga MT, Kuroda T, Nishizuka Y. Limited proteolysis of protein Zweerink HJ. Human cytotoxic lymphocyte granzyme B: its puri- kinase C subspecies by calcium-dependent neutral protease fication from granules and the characterization of substrate and (calpain). J Biol Chem 1989; 264: 4088–4092. inhibitor specificity. J Biol Chem 1991; 266: 98–103. 105 McGinnis KM, Whitton MM, Gnegy ME, Wang KKW. 129 Shi L, Kraut RP, Aebersold R, Greenberg AH. A natural killer cell Calcium/calmodulin-dependent protein kinase IV is cleaved by granule protein that induces DNA fragmentation and apoptosis. caspase-3 and calpain in SH-SY5Y human neuroblastoma cells J Exp Med 1992; 175: 553–566. undergoing apoptosis. J Biol Chem 1998; 273: 19993–20000. 130 Heusel JW, Wesselschmidt RL, Shresta S, Russell JH, Ley TJ. 106 Watanabe N, Vande Woude GF, Ikawa Y, Sagata N. Specific Cytotoxic lymphocytes require granzyme B for the rapid induc-

Leukemia Review DE Johnson 1703 tion of DNA fragmentation and apoptosis in allogeneic target 147 Boldin MP, Goncharov TM, Goltsev YV, Wallach D. Involve- cells. Cell 1994; 76: 977–987. ment of MACH, a novel MORT1/FADD-interacting protease, in 131 Froelich CJ, Orth K, Turbov J, Seth P, Gottlieb R, Babior B, Shah Fas/APO-1- and TNF receptor-induced cell death. Cell 1996; 85: GM, Bleackley RC, Dixit VM, Hanna W. New paradigm for lym- 803–815. phocyte granule-mediated cytotoxicity. Target cells bind and 148 Muzio M, Chinnaiyan AM, Kischkel FC, O’Rourke K, Shevch- internalize granzyme B, but an endosomolytic agent is necessary enko A, Ni J, Scaffidi C, Bretz JD, Zhang M, Gentz R, Mann for cytosolic delivery and subsequent apoptosis. J Biol Chem M, Krammer PH, Peter ME, Dixit VM. FLICE, a novel FADD- 1996; 271: 29073–29079. homologous ICE/CED-3-like protease, is recruited to the CD95 132 Jans DA, Jans P, Briggs LJ, Sutton V, Trapani JA. Nuclear transport (Fas/APO-1) death-inducing signaling complex. Cell 1996; 85: of granzyme B (fragmentin-2). J Biol Chem 1996; 271: 30781– 817–827. 30789. 149 Duan H, Orth K, Chinnaiyan AM, Poirier GG, Froelich CJ, He 133 Shi L, Mai S, Israels S, Browne K, Trapani JA, Greenberg AH. W-W, Dixit VM. ICE-LAP6, a novel member of the ICE/Ced-3 Granzyme B (GraB) autonomously crosses the gene family, is activated by the cytotoxic T cell protease gran- and perforin initiates apoptosis and GraB nuclear localization. J zyme B. J Biol Chem 1996; 271: 16720–16724. Exp Med 1997; 185: 853–866. 150 Fernandes-Alnemri T, Armstrong RC, Krebs J, Srinivasula SM, 134 Pinkoski MJ, Hobman M, Heibein JA, Tomaselli K, Li F, Seth P, Wang L, Bullrich F, Fritz LC, Trapani JA, Tomaselli KJ, Litwack Froelich CJ, Bleackley RC. Entry and trafficking of granzyme B G, Alnemri ES. In vitro activation of CPP32 and Mch3 by Mch4, in target cells during granzyme B-perforin-mediated apoptosis. a novel human apoptotic cysteine protease containing two Blood 1998; 92: 1044–1054. FADD-like domains. Proc Natl Acad Sci USA 1996; 93: 7464– 135 Pinkoski MJ, Heibein JA, Barry M, Bleackley RC. Nuclear translo- 7469. cation of granzyme B in target cell apoptosis. Cell Death Differ 151 Medema JP, Toes REM, Scaffidi C, Zheng TS, Flavell RA, Melief 2000; 7: 17–24. CJM, Peter ME, Offringa R, Krammer PH. Cleavage of FLICE 136 Pinkoski MJ, Winkler U, Hudig D, Bleackley RC. Binding of gran- (caspase-8) by granzyme B during cytotoxic T lymphocyte- zyme B in the nucleus of target cells. Recognition of an 80-kilod- induced apoptosis. Eur J Immunol 1997; 27: 3492–3498. alton protein. J Biol Chem 1996; 271: 10225–10229. 152 Van de Craen M, Van den brande I, Declercq W, Irmler M, Beya- 137 Trapani JA, Browne KA, Smyth MJ, Jans DA. Localization of gran- ert R, Tschopp J, Fiers W, Vandenabeele P. Cleavage of caspase zyme B in the nucleus. A putative role in the mechanism of cyto- family members by granzyme B: a comparative study in vitro. toxic lymphocyte-mediated apoptosis. J Biol Chem 1996; 271: Eur J Immunol 1997; 27: 1296–1299. 4127–4133. 153 Talanian RV, Yang XH, Turbov J, Seth P, Ghayur T, Casiano CA, 138 Darmon AJ, Nicholson DW, Bleackley RC. Activation of the Orth K, Froelich CJ. Granule-mediated killing: pathways for gran- apoptotic protease CPP32 by cytotoxic T-cell-derived granzyme zyme B-initiated apoptosis. J Exp Med 1997; 186: 1323–1331. B. Nature 1995; 377: 446–448. 154 Shi L, Chen G, MacDonald G, Bergeron L, Li H, Miura M, Rotello 139 Darmon AJ, Ley TJ, Nicholson DW, Bleackley RC. Cleavage of RJ, Miller DK, Li P, Seshadri T, Yuan J, Greenberg AH. Activation CPP32 by granzyme B represents a critical role for granzyme B of an interleukin 1 converting enzyme-dependent apoptosis path- in the induction of target cell DNA fragmentation. J Biol Chem way by granzyme B. Proc Natl Acad Sci USA 1996; 93: 1996; 271: 21709–21712. 11002–11007. 140 Martin SJ, Amarante-Mendes GP, Shi LF, Chuang TH, Casiano 155 Andrade F, Roy S, Nicholson D, Thornberry N, Rosen A, Casci- CA, O’Brien GA, Fitzgerald P, Tan EM, Bokoch GM, Greenberg ola-Rosen L. Granzyme B directly and efficiently cleaves several AH, Green DR. The cytotoxic cell protease granzyme B initiates downstream caspase substrates: implications for CTL-induced apoptosis in a cell-free system by proteolytic processing and acti- apoptosis. Immunity 1998; 8: 451–460. vation of the ICE/CED-3 family protease, CPP32, via a novel two- 156 Gershenfeld HK, Weissman IL. Cloning of a cDNA for a T cell- step mechanism. EMBO J 1996; 15: 2407–2416. specific serine protease from a cytotoxic T lymphocyte. Science 141 Quan LT, Tewari M, O’Rourke K, Dixit VM, Snipas SJ, Poirier 1986; 232: 854–858. GG, Ray C, Pickup DJ, Salvesen GS. Proteolytic activation of the 157 Masson D, Zamai M, Tschopp J. Identification of granzyme A cell death protease Yama/CPP32 by granzyme B. Proc Natl Acad isolated from cytotoxic T-lymphocyte-granules as one of the pro- Sci USA 1996; 93: 1972–1976. teases encoded by CTL-specific genes. FEBS Lett 1986; 208: 142 Fernandes-Alnemri T, Litwack G, Alnemri ES. Mch2, a new 84–88. member of the apoptotic Ced-3/Ice cysteine protease gene fam- 158 Ebnet K, Hausmann M, Lehmann-Grube F, Mullbacher A, Kopf ily. Cancer Res 1995; 55: 2737–2742. M, Lamers M, Simon MM. Granzyme A-deficient mice retain 143 Orth K, Chinnaiyan AM, Garg M, Froelich CJ, Dixit VM. The potent cell-mediated cytotoxicity. EMBO J 1995; 14: 4230–4239. CED-3/ICE-like protease Mch2 is activated during apoptosis and 159 Mullbacher A, Ebnet K, Blanden RV, Hla RT, Stehle T, Museteanu cleaves the death substrate lamin A. J Biol Chem 1996; 271: C, Simon MM. Granzyme A is critical for recovery of mice from 16443–16446. infection with the natural cytopathic viral pathogen, ectromelia. 144 Fernandes-Alnemri T, Takahashi A, Armstrong R, Krebs J, Fritz Proc Natl Acad Sci USA 1996; 93: 5783–5787. L, Tomaselli KJ, Wang L, Yu Z, Croce CM, Salvesen G, Earnshaw 160 Shresta S, Graubert TA, Thomas DA, Raptis SZ, Ley TJ. Granzyme WC, Litwack G, Alnemri ES. Mch3, a novel human apoptotic A initiates an alternative pathway for granule-mediated cysteine protease highly related to CPP32. Cancer Res 1995; 55: apoptosis. Immunity 1999; 10: 595–605. 6045–6052. 161 Hayes MP, Berrebi GA, Henkart PA. Induction of target cell DNA 145 Chinnnaiyan AM, Orth K, Hanna WL, Duan HJ, Poirier GG, Fro- release by the cytotoxic T lymphocyte granule protease gran- elich CJ, Dixit VM. Cytotoxic T cell-derived granzyme B activates zyme A. J Exp Med 1980; 170: 933–946. the apoptotic protease ICE-LAP3. Curr Biol 1996; 6: 897–899. 162 Beresford PJ, Xia Z, Greenberg AH, Lieberman J. Granzyme A 146 Gu Y, Sarnecki C, Fleming MA, Lippke JA, Bleackley RC, Su MS- loading induces rapid cytolysis and a novel form of DNA damage S. Processing and activation of CMH-1 by granzyme B. J Biol independently of caspase activation. Immunity 1999; 10: 585– Chem 1996; 271: 10816–10820. 594.

Leukemia