DOI QR코드

DOI QR Code

Identification of the novel substrates for caspase-6 in apoptosis using proteomic approaches

  • Cho, Jin Hwa (Medical Proteomics Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB)) ;
  • Lee, Phil Young (Medical Proteomics Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB)) ;
  • Son, Woo-Chan (Asan Institute for Life Sciences, Asan Medical Center) ;
  • Chi, Seung-Wook (Medical Proteomics Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB)) ;
  • Park, Byoung Chul (Medical Proteomics Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB)) ;
  • Kim, Jeong-Hoon (Medical Proteomics Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB)) ;
  • Park, Sung Goo (Medical Proteomics Research Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB))
  • Received : 2013.04.05
  • Accepted : 2013.05.09
  • Published : 2013.12.31

Abstract

Apoptosis, programmed cell death, is a process involved in the development and maintenance of cell homeostasis in multicellular organisms. It is typically accompanied by the activation of a class of cysteine proteases called caspases. Apoptotic caspases are classified into the initiator caspases and the executioner caspases, according to the stage of their action in apoptotic processes. Although caspase-3, a typical executioner caspase, has been studied for its mechanism and substrates, little is known of caspase-6, one of the executioner caspases. To understand the biological functions of caspase-6, we performed proteomics analyses, to seek for novel caspase-6 substrates, using recombinant caspase-6 and HepG2 extract. Consequently, 34 different candidate proteins were identified, through 2-dimensional electrophoresis/MALDI-TOF analyses. Of these identified proteins, 8 proteins were validated with in vitro and in vivo cleavage assay. Herein, we report that HAUSP, Kinesin5B, GEP100, SDCCAG3 and PARD3 are novel substrates for caspase-6 during apoptosis.

Keywords

INTRODUCTION

Apoptosis is a programmed cell death in multicellular organisms, employed for the development and maintenance of homeostasis, and removal of damaged cells (1). It has been demonstrated that caspases play a pivotal role in apoptosis (2). Caspases cleave their substrates after aspartic acid at P1 sites, by using cysteine residue in the catalytic site (3). P4-P1 residue in the target proteins determines the substrate specificity of caspases (4,5). In apoptosis, caspases are classified into initiator caspases and executioner caspases (6). Once the extrinsic death signal is given by the ligand-induced activation of death receptors, initiator caspase-8 and caspase-10 are activated by dimerization (7,8). The death signal is delivered by an internal factor, such as DNA damage, and the release of cytochrome c from the mitochondria induces the formation of the apoptosome. Initiator caspase-9 is recruited to the apoptosome complex, and activated by dimerization (9). In turn, this activated initiator caspases activate the executioner caspases. The activated executioner caspases cleave their specific target proteins (10). Executioner caspases include caspase-3, -7 and caspase-6. Among them, it has been well known that caspase-3 cleaves a large set of substrates, and plays a major role in apoptosis (11,12). Caspase-7, sharing the high sequence homology with caspase-3, recognizes the same cleavage site of caspase-3 (4,13), suggesting that caspase-3 and -7 have redundant roles (11). However, caspase-6 has somewhat unique roles as an executioner in apoptosis. Caspase-6 prefers leucine or valine at the P4 position, whereas caspase-3 and -7 prefer aspartate at the P4 position in target substrates (14,15). Therefore, caspase-6 has unique substrates that are not cleaved by either caspase-3 or caspase-7, such as lamin A/C (16), special AT-rich binding protein-1 (SATB1) (17), the neurodegenerative disease proteins Huntingtin (18), amyloid precursor protein (19), Presenilin 1, 2 (20), and DJ-1 (21). The most distinctive feature of caspase-6 is that it is activated by caspase-3 and -7 (22), and inversely activates caspase-3 and -7 (23). Also, caspase-6 acts as an initiator caspase, by processing initiator caspase-8 and -10 during apoptosis (22,24). Although the role of caspase-6 has been proposed in a number of reports, its substrates and its function are still unclear during apoptosis.

Thus, to better understand the biological roles of caspase-6 during apoptosis, we carried out proteomic analyses to seek for novel sets of caspase-6 substrates. In our present study, we report novel putative 34 substrates for caspase-6, and show that 5 of them, HAUSP, Kinesin5B, GEP100, SDCCAG3, and PARD3, are directly cleaved by caspase-6 during apoptosis.

 

RESULTS AND DISCUSSION

Screening of putative substrates for caspase-6 by proteomic approaches

To seek for novel caspase-6 substrates, we performed two-dimensional gel electrophoresis based proteomics analyses (Fig. 1A). HepG2, a human hepatoma cell line was used, since HepG2 cells have not been studied to screen for caspase substrates (14). To increase the possibility of finding more candidates in 2DE analyses, HepG2 cell extract was fractionated into cytosol, membrane and nucleus fractions, and each subcellular fraction was incubated with either recombinant active caspase-6 or catalytic mutant substituted 163-cysteine into serine in active site (inactive caspase-6). Then, incubated proteins were separated by isoelectric focusing and SDS-PAGE (Fig. 1A). The experiments were repeated three times, independently, to establish reproducibility. By comparing two-dimensional gel images of active caspase-6 and inactive caspase-6 mutant, appeared or disappeared protein spots were selected. Consequently, we found different 39 spots in cytosolic (Fig. 1B), 33 spots in membrane (Fig. 1C), and 62 spots in nucleus fraction (Fig. 1D). These protein spots were identified by MALDI-TOF- MS analysis. To obtain reliable results, proteins that have >95% confidence level in the Mascot program (Matrix Science) were selected. To determine whether the identified candidates were novel or not, we carried out a database search at The CASBAH (http://bioinf.gen.tcd.ie/casbah) and the CutDB, a proteolytic event database (http://cutdb.burnham.org), and a literature search at Pubmed. Finally, we obtained a list of novel candidates of caspase-6 substrates (Table 1). It is worth noting that the representative caspase-6 substrates, such as lamin A/C, vimentin, actin, tubulin, and ezrin, were identified in our screening system indicating that our screening method was functioning properly (Table 1).

HAUSP, Kinesin5B, GEP100, SDCCAG3 and PARD3 are directly cleaved by caspase-6 in vitro

In order to confirm whether the candidate proteins listed in Table 1 would be cleaved by caspase-6, we performed the in vitro cleavage assay. HepG2 cell extracts were incubated with either active caspase-6, or catalytic mutant caspase-6. The cleavage was detected by western blotting, using the specific antibodies. Eight candidate proteins (HAUSP, Kinesin 5B, GEP100, SDCCAG3, PARD3, CLK3, ADH4, and MDH1), of which commercial antibodies are available, were tested. Lamin A/C, a well known caspase-6 substrate, was used as a positive control. As a result, the level of HAUSP, Kinesin 5B, GEP100, SDCCAG3 and PARD3 reduced only when active caspase-6 was treated. This indicates that these proteins were cleaved directly by caspase-6, according to Lamin A/C cleavage (Fig. 2A). However, these cleavages were blocked in the presence of caspase-6 inhibitor, Z-VEID-FMK (Fig. 2A). Therefore, these results indicate that HAUSP, Kinesin5B, GEP100, SDCCAG3 and PARD3 are directly cleaved by caspase-6. On the other hand, CLK3, ADH4 and MDH1 were not cleaved by caspase-6, even though twice the amounts of caspase-6 were used (Fig. 2B), implying that CLK3, ADH4 and MDH1 are not direct substrates for caspase-6.

Fig. 1.(A) The workflow for screening to identify novel caspase-6 substrates by proteomics approaches. (B-D) 2-DE gel images showing the differentially displayed spots. (B) Cytosol fractions, (C) membrane fractions, or (D) nucleus fractions, treated with either active caspase-6 or inactive casspase-6, were analyzed using 2-DE. The experiments were repeated for three times, independently. The representative gel images are shown.

Table 1.aproteins scores are significant at 95% confidence level.

Candidate proteins were proteolytically processed by caspases during apoptosis

Then we performed the in vivo cleavage assay to verify whether the candidates would be cleaved in vivo during apoptosis. HeLa cells were treated with staurosporine (STS), which induces apoptosis by inhibiting cellular protein kinase (25). Apoptosis was confirmed by observing of PARP cleavage (Fig. 3A and 3B), which is a well-established apoptosis hallmark, and is also a caspase-3 and caspase-7 substrate (26,27). To examine the specificity of caspases, three different caspase inhibitors (Z-VEID-FMK; caspase-6 inhibitor, Z-DEVD-FMK; caspase-3/-7 inhibitor and Z-VAD-FMK; pan-caspase inhibitor) were treated together with STS. In STS stimulated HeLa cells, all candidate proteins (HAUSP, Kinesin5B, GEP100, SDCCAG3, and PARD3) were processed (Fig. 3A). In the presence of Z-VEID-FMK and STS, the cleavage of Kinesin5B and GEP100 was blocked, and recovered to the level of DMSO treated (Fig. 3A). Meanwhile, the cleavage of HAUSP, SDCCAG3, and PARD3 was inhibited slightly. Upon Z-DEVD-FMK and STS treatment, the cleavages of Kinesin5B, GEP100, and SDCCAG3 were inhibited completely, but HAUSP and PARD3 were still cleaved, but less than STS alone (Fig. 3A). Upon STS and Z-VAD-FMK treatment, the cleavage of all candidate proteins was inhibited completely (Fig. 3A). Accordingly, these data suggest that HAUSP, Kinesin5B, GEP100, SDCCAG3, and PARD3 are cleaved somewhat, not only by caspase-6, but also by other caspases, and their cleavages seem not to be mutually exclusive in vivo.

We also checked the rest of the candidates that were not cleaved directly by caspase-6, in the same way (Fig. 3B). CLK3 and ADH4 were proteolytically processed upon STS treatment, but these cleavages were slightly blocked by Z-VEID-FMK or Z-DEVD-FMK, and completely blocked by Z-VAD-FMK. Meanwhile the cleavage of MDH1 was almost completely inhibited by either Z-VEID-FMK or Z-DEVD-FMK. Taken together, CLK3, ADH4, and MDH1 are processed during apoptosis, but they are not direct substrates for caspase-6.

Fig. 2.Cleavage of caspase-6 substrate candidates was confirmed by in vitro cleavage assay. (A) HepG2 extract was incubated with active caspase-6 (10 U) or inactive capsase-6 (10 U) in the presence or absence of 30 μM Z-VEID-FMK. The reaction mixture was analyzed by immunoblotting with the specific antibodies against (A) HAUSP, Kinesin5B, GEP100, SDCCAG3, PARD3 and Lamin A/C. (B) HepG2 extract was incubated with increasing amount of active caspase-6 (5, 10, 15, 20 U) or inactive capsase-6 (20 U). The reaction mixture was analyzed by immunoblotting with the specific antibodies against CLK3, ADH4, MDH1, and Lamin A/C (Cl, cleaved Lamin A/C). Lamin A/C cleavage was used as a positive control. The asterisk indicates a nonspecific band. The representative blot of at least two independent experiments is shown. 1 U is the amount of enzyme required to cleave 1nmole of caspase substrate completely in 1 hr.

Among the identified caspase-6 substrates, HAUSP is a deubiquitinating enzyme that removes ubiquitin from p53 and MDM2 (28). HAUSP plays a role in cell survival, proliferation and apoptosis, through regulating the p53 level (28). It has been reported that HAUSP is processed in a caspase-dependent manner during thymocyte apoptosis (29). In our study, we demonstrate that HAUSP is directly processed by caspase-6 in human cells, the cervical cancer cell HeLa (Fig. 2A and 3A) and hepatocarcinoma cell HepG2 (data not shown), indicating that HAUSP is caspase-6 specific substrate, and is not cell type specific. Another identified caspase-6 substrate, Kinesin 5B is microtubule associated motor protein transporting vesicles along the microtubule (30). It has been reported that kinectin, kinesin receptor protein, is cleaved by caspase, and the transport system becomes destroyed during apoptosis (31). Based on our results, caspase-6 indeed cleaves kinesin 5B transporting cellular cargoes, as well as cytoskeletal protein, such as tubulin and actin, suggesting that caspase-6 might be involved in the disruption of intracellular vesicle transport process, by inactivating kinesin 5B.

In conclusion, we identified 34 putative caspase-6 substrates, through 2-dimesional electrophoresis/MALDI-TOF analyses. Furthermore, we confirmed that HAUSP, Kinesin 5B, GEP100, SDCCAG3 and PARD3 are directly cleaved by caspase-6 during apoptosis. These newly identified substrates seem to be involved in membrane trafficking, cell polarization, and p53 stability, suggesting that our results may provide a clue to understanding the roles of caspase-6 in apoptosis.

Fig. 3.Cleavage of caspase-6 substrate candidates was confirmed by in vivo cleavage assay. HeLa cells were treated with 1 μM STS for 24 hr in the presence or absence of the 100 μM caspase inhibitors (Z-VEID-FMK; caspase-6 inhibitor, Z-DEVD-FMK; caspase-3/-7 inhibitor, Z-VAD-FMK; pan-caspase inhibitor) pretreated to cells for 3 hr. After 24 hr, cells were harvested and lysed. Cell lysates were analyzed by immunoblotting with the specific antibodies against (A) HAUSP, Kinesin5B, GEP100, SDCCAG3, PARD3, Lamin A/C (Cl, cleaved Lamin A/C), and PARP (Cl, cleaved PARP); and (B) CLK3, ADH4, MDH1, Lamin A/C, and PARP. A coomassie blue stain of PVDF membrane was the loading control. The representative blot of at least two independent experiments is shown.

 

MATERIALS AND METHODS

Cell culture

HeLa and HepG2 were maintained in Dulbecco’s Modified Eagle’s Medium (DMEM, Gibco, USA), supplemented with 10% fetal bovine serum (FBS, Gibco, USA) in a humidified atmosphere of 5% CO2 in air, at 37℃. HepG2 cells were fractionated using Qproteome Cell Compartment Kit (QIAGEN, USA), according to the manufacture’s instruction.

Antibodies and reagents

Anti-PARP (#9542), anti-Lamin A/C (#2032), and anti-HAUSP (#3277) were purchased from Cell Signaling (USA). Anti-ADH4 (ab83819), anti-CLK3 (ab54683), anti-Kinesin5B (ab42492), anti-PARD3 (ab40769), and anti-SDCCAG3 (ab97666) were from Abcam (USA). Anti-GEP100 (G4798) was from Sigma. Anti- MDHC (sc-166879) was from Santa Cruz (USA). Staurosporine (#1048-1) was from BioVision (CA). Z-VEID-FMK (FMK006), Z-DEVD-FMK (FMK004), and Z-VAD-FMK (FMK001) were purchased from R&D System (USA).

Cloning and mutagenesis of caspase-6

Δ23 caspase-6, prodomain deleted mutant, was subcloned to pET21a(+), using BamH I and Xho I sites. This construct was mutated using the QuikChange Site-Directed Mutagenesis Kit (Stratagene, CA), according to the manufacture’s instructions, with the following oligonucleotides: Caspase-6 Forward C163S 5’-TTTATCATTCAGGCAAGTCGGGGAAACCAGCAC-3’, Caspase-6 Reverse C163S 5’-GTGCTGGTTTCCCCGACTTGCCTGAATGATAAA-3’. The constructs were confirmed by sequencing at the Solgent (Daejeon, Korea).

Expression and purification of recombinant caspase-6

Recombinant Δ23 caspase-6 and Δ23 caspase-6 C163S with C-terminal His-tag were expressed and purified from E. coli BL21 codon plus (DE3) RIL (stratagene, CA), using affinity chromatography. In brief, cells were lysed in the lysis buffer (50 mM NaH2PO4, 500 mM NaCl, pH 8.0) with 0.5% sarcosine and EDTA free protease inhibitor (Roche, Germany). The lysate was clarified by centrifuging at 13,000 rpm, 4℃ for 15min and the supernatant was incubated with Ni-NTA agarose beads (QIAGEN, USA). After extensive washing, bound proteins were eluted with the lysis buffer containing 250 mM imidazole. Protein concentration was determined by the Bradford (Bio-Rad, USA) protein assay.

2-DE analysis and MALDI-TOF-MS

The protein samples were dissolved in 9 M urea, 2 M thiourea, 4% CHAPS, 50 mM DTT, 0.003% bromophenol blue, and 2% IPG buffer pH3-10 (GE healthcare, USA). The dissolved samples were applied onto immobilized 18 cm, pH 3-10 gradient strips (GE healthcare, USA). Strips were rehydrated, and isoelectric focusing was performed in a stepwise fashion (0:01 h at 300 V; 1:30 h at 3,500 V; 13:00 hr at 3,500 V). After the focused strips were equilibrated with DTT and iodoacetamide equilibration solution for 15 min respectively, the second-dimensional gel electrophoresis was performed on 10% SDS-PAGE. The gels were silver-stained, using PlusOne Silver Staining Kit, Protein (GE healthcare, USA), according to the manufacturer’s instructions. For mass spectrometry analysis, silver stained proteins were excised from the gels. The spots were destained and digested with trypsin, and the peptides from the spot were analyzed by MALDI-TOF, at Yonsei Proteome Research Center (Seoul, Korea). The database search was performed at The CASBAH and CutDB. The CASBAH (The CAspase Substrate DataBAse Homepage) is an online database containing all of the reported mammalian caspase substrates (32). CutDB is an online proteolytic event database that provides the list of protease and corresponding substrates and cleavage sites (33).

In vitro cleavage assay

HepG2 cells were lysed using NP40 lysis buffer (137 mM NaCl, 20 mM Tris-HCl pH 8.0, 10% Glycerol, 1% NP40, 2 mM EDTA) containing protease inhibitor (Roche, Germany). Cell lysates were incubated with either recombinant Δ23 caspase-6 protein or Δ23 caspase-6 C163S protein, at 37℃ for 24 hr. Reactions were stopped by adding Laemmli buffer, and were boiled at 100℃ for 5 min.

References

  1. Ellis, R. E., Yuan, J. Y. and Horvitz, H. R. (1991) Mechanisms and functions of cell death. Annu. Rev. Cell Biol. 7, 663-698. https://doi.org/10.1146/annurev.cb.07.110191.003311
  2. Raff, M. C., Barres, B. A., Burne, J. F., Coles, H. S., Ishizaki, Y. and Jacobson, M. D. (1993) Programmed cell death and the control of cell survival: lessons from the nervous system. Science 262, 695-700. https://doi.org/10.1126/science.8235590
  3. Alnemri, E. S., Livingston, D. J., Nicholson, D. W., Salvesen, G., Thornberry. N. A., Wong, W. W. and Yuan, J. (1996) Human ICE/CED-3 protease nomenclature. Cell 87, 171. https://doi.org/10.1016/S0092-8674(00)81334-3
  4. Lavrik, I. N., Golks, A. and Krammer, P. H. (2005) Caspases: pharmacological manipulation of cell death. J. Clin. Invest. 115, 2665-2672. https://doi.org/10.1172/JCI26252
  5. Thornberry, N. A., Rano, T. A., Peterson, E. P., Rasper, D. M., Timkey, T., Garcia-Calvo, M., Houtzaqer, V. M., Nordstrom, P. A., Roy, S., Vaillancourt, J. P., Chapman, K. T. and Nicholson, D. W. (1997) A combinatorial approach defines specificities of members of the caspase family and granzyme B. Functional relationships established for key mediators of apoptosis. J. Biol. Chem. 272, 17907-17911. https://doi.org/10.1074/jbc.272.29.17907
  6. Nicholson, D. W. (1999) Caspase structure, proteolytic substrates, and function during apoptotic cell death. Cell Death Differ. 6, 1028-1042. https://doi.org/10.1038/sj.cdd.4400598
  7. Peter, M. E. and Krammer, P. H. (2003) The CD95 (APO-1/Fas) DISC and beyond. Cell Death Differ. 10, 26-35. https://doi.org/10.1038/sj.cdd.4401186
  8. Lee, E., Seo, J., Jeong, M., Lee, S. and Song, J. (2012) The roles of FADD in extrinsic apoptosis and necrosis. BMB Rep. 45, 496-508. https://doi.org/10.5483/BMBRep.2012.45.9.186
  9. Kwon, K. B., Kim, E. K., Shin, B. C., Seo, E. A., Yang, J. Y. and Ryu, D. G. (2003) Herba houttuyniae extract induces apoptotic death of human promyelocytic leukemia cells via caspase activation accompanied by dissipation of mitochondrial membrane potential and cytochrome c release. Exp. Mol. Med. 35, 91-97. https://doi.org/10.1038/emm.2003.13
  10. Thornberry, N. A. and Lazebnik, Y. (1998) Caspases: enemies within. Science 281, 1312-1316. https://doi.org/10.1126/science.281.5381.1312
  11. Lamkanfi, M. and Kanneqanti, T. D. (2010) Caspase-7: a protease involved in apoptosis and inflammation. Int. J. Biochem. Cell Biol. 42, 21-24. https://doi.org/10.1016/j.biocel.2009.09.013
  12. Lee, A. Y., Park, B. C., Jang, M., Cho, S., Lee, D. H., Lee, S. C., Myung, P. K. and Park, S. G. (2004). Identification of caspase-3 degradome by two-dimensional gel electrophoresis and matrix-assisted laser desorption/ionization-time of flight analysis. Proteomics 4, 3429-3436. https://doi.org/10.1002/pmic.200400979
  13. Jang, M., Park, B. C., Kang, S., Lee, D. H., Cho, S., Lee, S. C., Bae, K. H. and Park, S. G. (2008) Mining of caspase-7 substrates using a degradomic approach. Mol. Cells. 26, 152-157.
  14. Crawford, E. D. and Wells, J. A. (2011) Caspase substrates and cellular remodeling. Annu. Rev. Biochem. 80, 1055-1087. https://doi.org/10.1146/annurev-biochem-061809-121639
  15. Lamkanfi, M., Declercq, W., Kalai, M., Saelens, X. and Vandenabeele, P. (2002) Alice in caspase land. A phylogenetic analysis of caspases from worm to man. Cell Death Differ. 9, 358-361. https://doi.org/10.1038/sj.cdd.4400989
  16. Takahashi, A., Alnemri, E. S., Lazebnik, Y. A., Fernandes-Alnemri, T. Litwack, G., Moir, R. D., Goldman, R. D., Poirier, G. G., Kaufmann, S. H. and Earnshaw, W. C. (1996) Cleavage of lamin A by $Mch2\alpha$ but not CPP32: Multiple interleukin $1\beta$-converting enzyme-related proteases with distinct substrate recognition properties are active in apoptosis. Proc. Natl. Acad. Sci. U. S. A. 93, 8395-8400. https://doi.org/10.1073/pnas.93.16.8395
  17. Galande, S., Dickinson, L. A., Mian, I. S., Sikorska, M. and Kohwi-Shigematsu, T. (2001) SATB1 cleavage by caspase 6 disrupts PDZ domain-mediated dimerization, causing detachment from chromatin early in T-cell apoptosis. Mol. Cell Biol. 21, 5591-5604. https://doi.org/10.1128/MCB.21.16.5591-5604.2001
  18. Graham, R. K., Deng, Y., Slow, E. J., Haigh, B., Bissada, N., Lu, G., Pearson, J., Shehadeh, J., Bertram, L., Murphy, Z., Warby, S. C., Doty, C. N., Roy, S., Wellington, C. L., Leavitt, B. R., Raymond, L. A., Nicholson, D. W. and Hayden, M. R. (2006) Cleavage at the caspase-6 site is required for neuronal dysfunction and degeneration due to mutant huntingtin. Cell 125, 1179-1191. https://doi.org/10.1016/j.cell.2006.04.026
  19. Pellegrini, L., Passer, B. J., Tabaton, M., Ganjei, J. K. and D'Adamio, L. (1999) Alternative, non-secretase processing of Alzheimer's beta-amyloid precursor protein during apoptosis by caspase-6 and -8. J. Biol.Chem. 274, 21011-21016. https://doi.org/10.1074/jbc.274.30.21011
  20. van de Craen, M., de Jonghe, C., van den Brande, I., Declercq, W., van Gassen, G., van Criekinge, W., Vanderhoeven, I., Fiers, W., van Broeckhoven, C., Hendriks, L. and Vandenabeele, P. (1999) Identification of caspases that cleave presenilin-1 and presenilin-2. Five presenilin-1 (PS1) mutations do not alter the sensitivity of PS1 to caspases. FEBS Lett. 445, 149-154. https://doi.org/10.1016/S0014-5793(99)00108-8
  21. Giaime, E., Sunyach, C., Druon, C., Scarzello, S., Robert, G., Grosso, S., Auberger, P., Goldberg, M. S., Shen, J., Heutink, P., Pouyssegur, J., Paqes, G., Checler, F. and Alves da Costa, C. (2010) Loss of function of DJ-1 triggered by Parkinson's disease-associated mutation is due to proteolytic resistance to caspase-6. Cell Death Differ. 17, 158-169. https://doi.org/10.1038/cdd.2009.116
  22. Inoue, S., Browne, G., Melino, G. and Cohen, G. M. (2009) Ordering of caspases in cells undergoing apoptosis by the intrinsic pathway. Cell Death Differ. 16, 1053-1061. https://doi.org/10.1038/cdd.2009.29
  23. Allsopp, T. E., McLuckie, J., Kerr, L. E., Macleod, M., Sharkey, J. and Kelly, J. S. (2000) Caspase 6 activity initiates caspase 3 activation in cerebellar granule cell apoptosis. Cell Death Differ. 7, 984-993. https://doi.org/10.1038/sj.cdd.4400733
  24. Cowling, V. and Downward, J. (2002) Caspase-6 is the direct activator of caspase-8 in the cytochrome c-induced apoptosis pathway: absolute requirement for removal of caspase-6 prodomain. Cell Death Differ. 9, 1046-1056. https://doi.org/10.1038/sj.cdd.4401065
  25. Son, Y. K., Hong, D. H., Kim, D., Firth, A. L. and Park, W. S. (2011) Direct effect of protein kinase C inhibitors on cardiovascular ion channels. BMB Rep. 44, 559-565. https://doi.org/10.5483/BMBRep.2011.44.9.559
  26. Slee, E. A., Adrain, C. and Martin, S. J. (2001) Executioner caspase-3, -6, and -7 perform distinct, non-redundant roles during the demolition phase of apoptosis. J. Biol. Chem. 276, 7320-7326. https://doi.org/10.1074/jbc.M008363200
  27. Germain, M., Affar, E. B., D'Amours, D., Dixit, V. M., Salvesen, G. S. and Poirier, G. G. (1999) Cleavage of automodified poly(ADP-ribose) polymerase during apoptosis. Evidence for involvement of caspase-7. J. Biol. Chem. 274, 28379-28384. https://doi.org/10.1074/jbc.274.40.28379
  28. Li, M., Chen, D., Shiloh, A., Lou, J., Nikolaev, A. Y., Qin, J. and Gu, W. (2002) Deubiquitination of p53 by HAUSP is an important pathway for p53 stabilization. Nature 416, 648-653. https://doi.org/10.1038/nature737
  29. Vugmeyster, Y., Borodovsky, A., Maurice, M. M., Maehr, R., Furman, M. H. and Ploegh, H. L. (2002) The ubiquitin-proteosome pathway in thymocyte apoptosis: caspase-dependent processing of the deubiquitinating enzyme USP (HAUSP). Mol. Immunol. 39, 431-441. https://doi.org/10.1016/S0161-5890(02)00123-2
  30. Brady, S. T. (1985) A novel brain ATPase with properties expected for the fast axonal transport motor. Nature 317, 73-75. https://doi.org/10.1038/317073a0
  31. Machleidt, T., Geller, P., Schwandner, R., Scherer, G. and Kroenke, M. (1998) Caspase 7-induced cleavage of kinectin in apoptotic cells. FEBS Lett. 436, 51-54. https://doi.org/10.1016/S0014-5793(98)01095-3
  32. Luthi, A. U. and Martin, S. J. (2007) The CASBAH: a searchable database of caspase substrates. Cell Death. Differ. 14, 641-650. https://doi.org/10.1038/sj.cdd.4402103
  33. Igarashi, Y., Eroshkin, A., Gramatikova, S., Gramatikoff, K., Zhang, Y., Smith, J. W., Osterman, A. L. and Godzik, A. (2007) CutDB: a proteolytic event database. Nucleic Acids Res. 35, D546-549. https://doi.org/10.1093/nar/gkl813

Cited by

  1. Crosstalk between Autophagy and Apoptosis: Potential and Emerging Therapeutic Targets for Cardiac Diseases vol.17, pp.3, 2016, https://doi.org/10.3390/ijms17030332
  2. Identification of cleavage of NS5A of C-strain classical swine fever virus vol.162, pp.2, 2017, https://doi.org/10.1007/s00705-016-3117-z
  3. Mutation of the lbp-5 gene alters metabolic output in Caenorhabditis elegans vol.47, pp.1, 2014, https://doi.org/10.5483/BMBRep.2014.47.1.086
  4. Ginsenoside-Rp1-induced apolipoprotein A-1 expression in the LoVo human colon cancer cell line vol.38, pp.4, 2014, https://doi.org/10.1016/j.jgr.2014.06.003
  5. Proteolysis to Identify Protease Substrates: Cleave to Decipher vol.18, pp.13, 2018, https://doi.org/10.1002/pmic.201800011
  6. Differential susceptibility of striatal, hippocampal and cortical neurons to Caspase-6 vol.25, pp.7, 2018, https://doi.org/10.1038/s41418-017-0043-x