Cytoskeletal keratin 18 (K18) undergoes caspase-mediated digestion during apoptosis, which leads to dramatic disassembly of keratin filaments. We studied the significance of K18 caspase digestion in a mouse model and generated transgenic mice expressing the human K18 caspase digestion-resistant double-mutant K18-D238/397E in a mouse (m) K18-null background, and compared their response to injury mediated by administration of antibody against tumor necrosis factor receptor superfamily member 6 (Fas), anti-FasAb. Notably, K18-D238/397E;mK18-null mice were significantly more resistant to anti-FasAb-induced injury as compared with K18-WT;mK18-null mice (23% vs 57% lethality, respectively; P<0.001). The same applied when the toxin microcystin-LR (MLR) was used to induce liver injury, i.e. lethality of K18-D238/397E;mK18-null mice in response to MLR treatment was reduced compared with the control mouse strain. The lesser rate of apoptosis in K18-D238/397E;mK18-null livers is associated with delayed degradation and, thus, sustained activation of cell-survival-related protein kinases, including stress-activated protein kinases and the NF-κB transcription factor, up to 6-8 h after administration of anti-FasAb. However, activation of the kinases and NF-κB in K18-WT-reconstituted livers decreases dramatically 8 h after anti-FasAb administration. In addition, the D238/397E double-mutation results in prolonged stability of K18 protein in transfected cells and transgenic livers. Therefore, our results show that the caspase digestion-resistant K18 helps to maintain keratin filament organization and delays apoptosis, thereby resulting in protection from liver injury.

Keratin 8 and keratin 18 (KRT8 and KRT18, respectively; hereafter referred to as K8/K18) in the liver are a pair of intermediate filament proteins that consist of a central α-helical rod domain linked to the non-α-helical N-terminal head and C-terminal tail domains (Herrmann et al., 2009; Omary et al., 2009). They form obligate noncovalent heteropolymers that assemble into filaments and generate the cytoplasmic filament network that maintains cellular integrity (Coulombe et al., 2009). Keratin filaments are highly dynamic structures that can disassemble into nonfilamentous particles in mitotic cells or under various stresses (Eriksson et al., 2009).

Apoptosis occurs in several tissues during development and disease to remove harmful and unneeded cells; its distinct morphological changes include cytoskeletal and nuclear envelope disruption, chromatin compaction, cell shrinkage, cell fragmentation and formation of apoptotic bodies (Earnshaw, 1995; MacFarlane et al., 2000). Caspases, which are cysteine-dependent aspartate (Asp) proteases, cleave their substrates at Asp residues and play essential roles in apoptosis. There are two classes of caspases: initiator caspases that include caspases 2, 8, 9 and 10, and effector caspases that include caspases 3, 6 and 7 (Riedl and Shi, 2004). Initiators cleave and activate the effectors, which in turn cleave other substrates to promote apoptosis.

Various intermediate filament proteins, such as lamins (Lazebnik et al., 1995; Rao et al., 1996; Ruchaud et al., 2002), desmin (Chen et al., 2003), vimentin (Byun et al., 2001) and keratins (Caulín et al., 1997; Ku et al., 1997, 2016; Ku and Omary, 2001) are known caspase substrates. Caspase-dependent digestion of intermediate filaments leads to breakdown of the filament network and may facilitate morphological changes in cells that are associated with apoptosis. Resistance of caspase cleavage is essential in protecting the cells from apoptosis; for example, nuclear lamins − which are main elements of nuclear lamina − have caspase cleavage sites, i.e. VEID230 in lamin A and VEVD231 in lamin B. Interrupting the caspase-dependent cleavage of lamins delays nuclear events during apoptosis, such as nuclear shrinkage, DNA fragmentation and onset of apoptosis (Lazebnik et al., 1995; Rao et al., 1996; Ruchaud et al., 2002). In the case of the muscle-specific intermediate filament protein desmin, VEMD263 is a caspase-6 cleavage site (Chen et al., 2003). Cells transfected with caspase-resistant desmin D263E mutant manifest alleviated apoptosis as compared with wild-type (WT) desmin-transfected cells (Chen et al., 2003). Consistent with these findings, desmin that cannot be cleaved by caspase protects cardiomyocytes from apoptosis and improves heart function during TNFα-mediated stress (Panagopoulou et al., 2008).

K18 has two caspase cleavage sites, VEVD238 and DALD397, and is digested during apoptosis. By contrast, K8 does not have the caspase cleavage consensus sequences and is, therefore, relatively resistant to apoptotic degradation (Caulín et al., 1997; Ku et al., 1997; Leers et al., 1999). VEVD238 or similar sequences are found in the rod domain of other intermediate filament proteins, whereas DALD397 is found only in the K18 tail domain (Ku and Omary, 2001). Apoptotic signals activate caspase-9, which is responsible for DALD397cleavage as an early event (Ku and Omary, 2001; Riedl and Shi, 2004). The breakdown of the keratin network into large aggregates is due to VEVD238 cleavage by activated caspase-6 in a later stage of apoptosis, with the consequent movement of the large keratin aggregates into cytoplasmic blebs (Schutte et al., 2004). Mutations of two Asp residues in K18 (D238/D397) to Glu (K18-D238/397E) renders K18 resistant to caspase-mediated digestion under cell culture conditions (Ku and Omary, 2001). Recently, it has been reported that the human K18-D238/397E double mutant, when overexpressed as a transgene in mice that retain endogenous mouse WT K18, results in keratin hypo-phosphorylation, reorganization and increased susceptibility to necrosis (Weerasinghe et al., 2014). However, the function of uncleavable K18 without endogenous mouse WT K18 has, so far, not been investigated in mice.

We, therefore, studied here the significance of K18 caspase cleavage by generating K18-D238/397E mice in a K18-null background (K18-D238/397E;mK18-null) by crossbreeding K18-D238/397E mice with K18-null mice. The K18-D238/397E;mK18-null mice showed remarkable resistance to liver injury when compared with control mice, based on mouse mortality, histopathological and biochemical analysis, terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) assay and caspase activation. In addition, the non-cleavage form of K18 is more stable than WT K18 when tested in cells treated with cycloheximide or in transgenic mice treated with antibody against the tumor necrosis factor receptor superfamily member 6 (Fas), hereafter referred to as anti-FasAb. Therefore, caspase cleavage-resistant K18 mutation enhances resistance to keratin network disruption under stress, thereby providing protection of liver injury induced by treatment with anti-FasAb.

In transgenic mice that express endogenous mouse K18, expression of K18-D238/397E does not impact on anti-FasAb-mediated liver injury

To investigate the biological significance of K18 cleavage at D238 and D397 during apoptosis (Fig. 1A), we used transgenic mice that overexpress human K18-D238/397E in an FVB/N mouse-strain background, which contains endogenous mouse K18. Expression of K18-D238/397E in mouse liver was confirmed by the lack of the two apoptotic fragments of human K18 (p43 and p29) that are observed in transgenic liver overexpressing human K18-WT as the result of anti-FasAb-mediated apoptosis (Fig. 1B). The mice did not show abnormal phenotypes under basal conditions (not shown). We then compared the mouse lethality of nontransgenic FVB/N, K18-WT and K18-D238/397E mice after anti-FasAb administration to induce apoptosis in the liver. After anti-FasAb injection, the death of mice starts after 8 h but no further death is noted beyond 50 h (Fig. 1C). Mouse mortality was slightly higher in K18-D238/397E mice when compared with nontransgenic FVB/N and K18-WT mice (53% compared with 44% and 42%, respectively; Fig. 1D). The trend of these findings parallels a previous study, which demonstrated that the K18-D238/397E mutation enhanced the susceptibility to anti-FasAb-mediated liver injury (Weerasinghe et al., 2014). However, in our study, the difference of lethality between the K18-D238/397E mice and control mice was not statistically significant (P>0.58) (Fig. 1D).

Fig. 1.

Expression of K18-D238/397E in nontransgenic FVB/N background has no obvious effect on progression of apoptosis induced by anti-FasAb. (A) Schematic representation of K18 caspase cleavage sites. Similar to other intermediate filament proteins, K18 has the structural motifs that consist of a central α-helical domain that is flanked by non-helical head and tail domains (not drawn to scale). Main phosphorylation and glycosylation sites are in the head domain whereas two caspase cleavage sites are in the rod and tail domains (Snider and Omary, 2014). The cleavage site in rod domain (VEVD238) is highly conserved in intermediate filament proteins, but the site in tail domain (DALD397) is present only in K18. (B) Mice were injected intraperitoneally with anti-FasAb (0.15 mg/kg body weight) to induce apoptosis or with PBS as a control. Livers were isolated after 6 h, and total liver lysates were prepared and immunoblotted with the indicated antibodies. Human K18 protein (hK18) in transgenic liver of transgenic mice was detected by immunoblotting with antibody specifically recognizing human K18. Expression of K18-D238/397E in the liver of mutant mice was verified by the lack of the two apoptotic fragments of human K18 (p43 and p29) that we usually observed in K18-WT liver after injection with anti-FasAb. Coomassie Blue staining is included to show equal protein loading. Each lane represents the analysis of one individual liver. Note that the levels of cleaved caspase 7 were almost the same in WT and D238/397E livers, which is consistent with similar progression of apoptosis. mK8, mouse keratin 8. (C,D) Nontransgenic FVB/N mice and transgenic mice overexpressing K18-WT or D238/397E were injected with FasAb as described in panel B. The mice were observed hourly for the first 12 h and every 8-10 h for 3 days. No deaths were detected after 50 h. Survival curves (C) and mortality (D) are shown. The increased lethality of K18-D238/397E mice compared with nontransgenic FVB/N and K18-WT mice was not statistically significant (P>0.58). P>0.55 when comparing K18-D238/397E with K18-WT.

Fig. 1.

Expression of K18-D238/397E in nontransgenic FVB/N background has no obvious effect on progression of apoptosis induced by anti-FasAb. (A) Schematic representation of K18 caspase cleavage sites. Similar to other intermediate filament proteins, K18 has the structural motifs that consist of a central α-helical domain that is flanked by non-helical head and tail domains (not drawn to scale). Main phosphorylation and glycosylation sites are in the head domain whereas two caspase cleavage sites are in the rod and tail domains (Snider and Omary, 2014). The cleavage site in rod domain (VEVD238) is highly conserved in intermediate filament proteins, but the site in tail domain (DALD397) is present only in K18. (B) Mice were injected intraperitoneally with anti-FasAb (0.15 mg/kg body weight) to induce apoptosis or with PBS as a control. Livers were isolated after 6 h, and total liver lysates were prepared and immunoblotted with the indicated antibodies. Human K18 protein (hK18) in transgenic liver of transgenic mice was detected by immunoblotting with antibody specifically recognizing human K18. Expression of K18-D238/397E in the liver of mutant mice was verified by the lack of the two apoptotic fragments of human K18 (p43 and p29) that we usually observed in K18-WT liver after injection with anti-FasAb. Coomassie Blue staining is included to show equal protein loading. Each lane represents the analysis of one individual liver. Note that the levels of cleaved caspase 7 were almost the same in WT and D238/397E livers, which is consistent with similar progression of apoptosis. mK8, mouse keratin 8. (C,D) Nontransgenic FVB/N mice and transgenic mice overexpressing K18-WT or D238/397E were injected with FasAb as described in panel B. The mice were observed hourly for the first 12 h and every 8-10 h for 3 days. No deaths were detected after 50 h. Survival curves (C) and mortality (D) are shown. The increased lethality of K18-D238/397E mice compared with nontransgenic FVB/N and K18-WT mice was not statistically significant (P>0.58). P>0.55 when comparing K18-D238/397E with K18-WT.

The K18-D238/397E mutation protects from liver injury in transgenic mice that lack endogenous mouse K18

To generate K18-D238/397E mice that lack endogenous mouse K18 (K18-D238/397E;mK18-null), we crossbred K18-D238/397E mice with K18-null mice. As a control, we also generated K18-WT;mK18-null mice that expresses human K18-WT but not endogenous mouse K18-WT. No mouse K18 was observed in K18-D238/397E;mK18-null or K18-WT;mK18-null mice livers, as shown in Coomassie Blue-stained high-salt-extracted samples − which generate a highly enriched keratin fraction (Ku et al., 2004) − and immunoblots of total liver lysates (Fig. 2A). Given that the genetic background of the generated K18-WT;mK18-null mice is K18-null, we first tested the susceptibility of K18-null mice to anti-FasAb-induced liver injury. The mortality is dramatically increased in K18-null mice when compared with the nontransgenic FVB/N mouse (94% vs 56%, P<0.04; Fig. 2B,E). This enhanced mortality is probably due to the lack of a keratin filament network in liver of K18-null mice as described previously (Magin et al., 1998). The increased mortality of K18-null mice is alleviated by expression of human K18 WT in K18-null mice (K18-WT;mK18-null), which is comparable to the lethality of nontransgenic FVB/N mice (57% vs 56%) (Fig. 2E). These findings indicate that the reintroduction of human K18-WT rescues K18-null mice from liver injury.

Fig. 2.

Expression of K18-D238/397E in mK18-null background protects transgenic mice from apoptosis induced by anti-FasAb or MLR. (A) K18-WT or D238/397E mice without mK18 were analyzed as described in Fig. 1, panel B. Notice that mK18 is not detected in K18-WT or D238/397E mice and the cleaved caspase 7 levels are lower in D238/397E livers as compared with WT livers after administration of anti-FasAb. This is consistent with D238/397E protection from apoptosis induced by anti-FasAb. (B-D) Survival curves. The indicated mice (9−52 mice, age- and sex-matched per genotype in panel E) were injected with anti-FasAb or microcystin-LR to induce liver damage. Mice were monitored hourly for 12 h, and then every 10 h for 50 h. No deaths were observed beyond 50 h. (E) Summary of mouse mortality data.

Fig. 2.

Expression of K18-D238/397E in mK18-null background protects transgenic mice from apoptosis induced by anti-FasAb or MLR. (A) K18-WT or D238/397E mice without mK18 were analyzed as described in Fig. 1, panel B. Notice that mK18 is not detected in K18-WT or D238/397E mice and the cleaved caspase 7 levels are lower in D238/397E livers as compared with WT livers after administration of anti-FasAb. This is consistent with D238/397E protection from apoptosis induced by anti-FasAb. (B-D) Survival curves. The indicated mice (9−52 mice, age- and sex-matched per genotype in panel E) were injected with anti-FasAb or microcystin-LR to induce liver damage. Mice were monitored hourly for 12 h, and then every 10 h for 50 h. No deaths were observed beyond 50 h. (E) Summary of mouse mortality data.

We then compared the susceptibility to liver injury mediated by anti-FasAb of K18-D238/397E;mK18-null mice with that of K18-WT;mK18-null mice and found that mortality is markedly reduced in K18-D238/397E;mK18-null mice (23% vs 57%, P<0.001) (Fig. 2C,E). Similar to the pattern of mortality after anti-FasAb administration, we also observed a reduced rate of mortality of the mice after injection of microcystin-LR (MLR) (Fig. 2D,E). The phosphatase inhibitor MLR is a known hepatotoxin (Jochimsen et al., 1998; Ku and Omary, 2006). These results clearly demonstrate that expression of K18-D238/397E in K18-null mice protects the mice from liver injury.

Mutation of K18-D238/397E helps to maintain cell integrity during apoptotic liver damage

Liver histology was carried out to determine the extent of tissue damage. Liver sections were prepared 6 h post treatment with anti-FasAb or vehicle (PBS). We observed no tissue damage in any mice injected with vehicle only (Fig. 3A). Administration of anti-FasAb caused severe hemorrhage after 6 h in liver of K18-WT;mK18-null mice, whereas the histological change was significantly attenuated in that of K18-D238/397E;mK18-null mice (Fig. 3A). These histological findings were supported by significantly lower levels of alanine transaminase (ALT) in K18-D238/397E;mK18-null serum (Fig. 3B). Given that involvement of apoptotic pathways is the dominant feature in anti-FasAb-mediated liver injury (Ogasawara et al., 1993), we compared the extent of apoptotic cell death by using a TUNEL assay. Neither group of mice injected with vehicle (PBS) alone showed TUNEL-positive apoptotic cells (Fig. 3C, left panels). However, after treatment with anti-FasAb, high numbers of apoptotic cells were observed in liver of K18-WT;mK18-null mice, whereas significantly fewer apoptotic cells were detected in that of K18-D238/397E;mK18-null mice (Fig. 3C, right panels). Thus, the K18-D238/397E mutation in livers of mK18-null mice protects them from liver apoptosis mediated by anti-FasAb.

Fig. 3.

Comparison of liver injury mediated by anti-FasAb in K18-WT and K18-D238/397E mice in K18-null background. Liver and blood obtained from the indicated mice, 6 h after intraperitoneal injection of anti-FasAb (+) or PBS (−). (A) Histologic analysis. The livers were fixed in 10% formalin, sectioned and stained with hematoxylin/eosin. Note that the mild hemorrhage (‘H’, highlighted by arrows) in K18-D238/397E as compared with K8-WT liver after administration of anti-FasAb. (B) After mice were killed through CO2 inhalation, their blood was collected by intra-cardiac puncture and the level of alanine transaminase (ALT) quantified [units per liter (U/L) as mean±s.e.m.; *P<0.0001]. (C,D) TUNEL assay and immunofluorescence staining. Isolated liver was frozen in OCT-filled cryomold and then sectioned. The liver sections were used for TUNEL analysis in order to detect (C) apoptotic cells (dark brown) and non-apoptotic cells (green) or (D) were stained with antibody against K18 (red). The number of TUNEL-positive cells from liver sections of mice injected with anti-FasAb was counted in four fields. Data are represented as the mean±s.e.m., **P<0.01. Note that, after injection of anti-FasAb, cells are less apoptotic and keratin filaments only mildly damaged in liver from D238/397E mice when compared with liver from WT mice. Scale bars: 200 μm (A,C), 100 μm (Da), 40 μm in (Da′).

Fig. 3.

Comparison of liver injury mediated by anti-FasAb in K18-WT and K18-D238/397E mice in K18-null background. Liver and blood obtained from the indicated mice, 6 h after intraperitoneal injection of anti-FasAb (+) or PBS (−). (A) Histologic analysis. The livers were fixed in 10% formalin, sectioned and stained with hematoxylin/eosin. Note that the mild hemorrhage (‘H’, highlighted by arrows) in K18-D238/397E as compared with K8-WT liver after administration of anti-FasAb. (B) After mice were killed through CO2 inhalation, their blood was collected by intra-cardiac puncture and the level of alanine transaminase (ALT) quantified [units per liter (U/L) as mean±s.e.m.; *P<0.0001]. (C,D) TUNEL assay and immunofluorescence staining. Isolated liver was frozen in OCT-filled cryomold and then sectioned. The liver sections were used for TUNEL analysis in order to detect (C) apoptotic cells (dark brown) and non-apoptotic cells (green) or (D) were stained with antibody against K18 (red). The number of TUNEL-positive cells from liver sections of mice injected with anti-FasAb was counted in four fields. Data are represented as the mean±s.e.m., **P<0.01. Note that, after injection of anti-FasAb, cells are less apoptotic and keratin filaments only mildly damaged in liver from D238/397E mice when compared with liver from WT mice. Scale bars: 200 μm (A,C), 100 μm (Da), 40 μm in (Da′).

Given the keratin filament network is crucial in maintaining cell integrity (Coulombe et al., 2009; Omary et al., 2009), we examined the extent of perturbation in the keratin network by keratin immunostaining of the liver sections (Fig. 3D) and the primary cultured hepatocytes (Fig. S1). Anti-FasAb treatment caused a more-prominent collapse of the keratin network in K18-WT;mK18-null hepatocytes than in K18-D238/397E;mK18-null hepatocytes. Collectively, these findings indicate that the caspase cleavage-resistant mutation of K18 helps to maintain hepatocyte integrity and protects liver of transgenic mice from apoptosis.

Effect of the K18-D238/397E mutation on protein stability and apoptosis

The protective role of K18-D238/397E mutation was further confirmed by immunoblotting, which shows markedly low levels of cleaved caspase 7 in most of the tested liver from K18-D238/397E;mK18-null mice as compared with that of K18-WT;mK18-null mice after injection with anti-FasAb (Fig. 4A). Notably, one of four livers of K18-D238/397E;mK18-null mice had a high level of cleaved caspase 7 that was probably caused by variation between individual mice as shown by the 23% mortality of K18-D238/397E; mK18-null mice in Fig. 2E. Overall, immunoblotting of cleaved caspase 7 suggested that one of the four K18-D238/397E mice underwent severe apoptosis whereas all four K18 WT mice show signs of apoptosis in their liver.

Fig. 4.

K18-D238/397E in an mK18-null background delays the onset of liver apoptosis induced by injection of anti-FasAb. (A) Livers from K18-WT and K18-D238/397E mice were harvested at 6 or 8 h after injection of anti-FasAb at 0.15 mg/kg body weight (+) or PBS (−), homogenized and solubilized in 2% SDS-containing sample buffer to prepare total liver lysates. Lysates were analyzed in SDS-PAGE and immunoblotted with indicated antibodies. Each lane represents analysis of one individual liver. Cleaved caspase 7 was detected at 8 h in D238/397E liver after treatment with anti-FasAb whereas it was already present at 6 h in WT anti-FasAb-treated liver, implying delayed apoptosis in D238/397E liver. In addition, a dramatic reduction in phosphorylation of protein kinases and NF-κB accompanied with a decrease in their protein levels was observed in the WT liver at 8 h after injection of anti-FasAb, probably because of rapid protein degradation during apoptosis. (B) Primary hepatocytes were isolated from K18-D238/397;mK18-null mice or K18-WT;mK18-null mice by liver perfusion and treated without or with FasAb (+, 0.5 μg/ml; +++, 1.5 μg/ml) for 6, 12 or 24 h. Total lysates were analyzed by SDS-PAGE and immunoblotted with indicated antibodies. (C) Primary hepatocytes as described for B were treated with the protein synthesis inhibitor cycloheximide (100 μg/ml) for the indicated times. Total lysates were immunoblotted with indicated antibodies. Hsc70 was used as a loading control (A-C).

Fig. 4.

K18-D238/397E in an mK18-null background delays the onset of liver apoptosis induced by injection of anti-FasAb. (A) Livers from K18-WT and K18-D238/397E mice were harvested at 6 or 8 h after injection of anti-FasAb at 0.15 mg/kg body weight (+) or PBS (−), homogenized and solubilized in 2% SDS-containing sample buffer to prepare total liver lysates. Lysates were analyzed in SDS-PAGE and immunoblotted with indicated antibodies. Each lane represents analysis of one individual liver. Cleaved caspase 7 was detected at 8 h in D238/397E liver after treatment with anti-FasAb whereas it was already present at 6 h in WT anti-FasAb-treated liver, implying delayed apoptosis in D238/397E liver. In addition, a dramatic reduction in phosphorylation of protein kinases and NF-κB accompanied with a decrease in their protein levels was observed in the WT liver at 8 h after injection of anti-FasAb, probably because of rapid protein degradation during apoptosis. (B) Primary hepatocytes were isolated from K18-D238/397;mK18-null mice or K18-WT;mK18-null mice by liver perfusion and treated without or with FasAb (+, 0.5 μg/ml; +++, 1.5 μg/ml) for 6, 12 or 24 h. Total lysates were analyzed by SDS-PAGE and immunoblotted with indicated antibodies. (C) Primary hepatocytes as described for B were treated with the protein synthesis inhibitor cycloheximide (100 μg/ml) for the indicated times. Total lysates were immunoblotted with indicated antibodies. Hsc70 was used as a loading control (A-C).

We examined the phosphorylation and expression of some apoptosis-related proteins−namely Akt, p42MAPK and p44MAPK (also known as MAPK1 and MAPK3, respectively), p90RSK (also known as RPS6KA1), p38MAPK (also known as MAPK14), JNK1 (also known as MAPK8), JNK2 (also known as MAPK9), NF-κB in anti-FasAb-treated livers. The phosphorylation and protein levels of NF-κB and the tested protein kinases were significantly increased and then were maintained in livers of K18-D238/397E mice. However, in livers of K18-WT mice, they had rapidly diminished 8 h after treatment with anti-FasAb (Fig. 4A), which is likely to be related to rapid protein degradation due to massive apoptosis in livers of K18-WT mice. Nonetheless, the reduced phosphorylation of proteins but not the overall reduced levels of proteins, might also have been involved in the substantially increased levels of apoptosis in the liver of K18 WT mice. For example, the protein levels of JNK1/2 8 h after treatment with anti-FasAb are similar in livers of K18-WT and K18-D238/397E mice, whereas levels of phosphorylated JNK1/2 in livers of K18-WT mice is more dramatically reduced when compared with those in livers of K18-D238/397E mice (Fig. 4A). In addition, other apoptosis-associated proteins, such as the Fas-associated protein with death domain (FADD) and Fas itself, are barely detectable in livers of K18 WT mice after stimulation with anti-FasAb, whereas levels of actin and Hsc70 are maintained (Fig. 4A). This might be due to the different accessibility of caspases, depending on their substrates. Specifically, actin forms a filament network and, thus, might be relatively stable under certain apoptotic conditions at which other cytosolic proteins (such as protein kinases described in Fig. 4A) are degraded. HSC70 (known as HSPA8) protein is also stable under apoptotic conditions and is used as a loading control protein.

We investigated the significance of specific protein degradation under apoptosis by using an ex vivo primary culture system. Primary hepatocytes were isolated from mouse liver that had been perfused and were treated with anti-FasAb. We then assessed the level of apoptosis and the amount of signaling proteins in both K18 WT and K18-D238/397E hepatocytes. Consistent with the protective role of K18-D238/397E mutation in vivo (Fig. 4A), we observed lower levels of cleaved caspase-7 and cleaved PARP in primary cultured K18-D238/397E hepatocytes as compared with K18-WT hepatocytes after treatment with anti-FasAb; levels of cleaved caspase-3 and caspase-8 were also reduced, albeit to a lesser extend (Fig. 4B). Notably, levels of the cell survival factor NF-κB are more stably maintained in both K18-D238/397E livers in vivo (Fig. 4A) and K18-D238/397E hepatocytes ex vivo (Fig. 4B). Indeed, NF-κB seemed more stable in K18-D238/397E hepatocytes compared with WT hepatocytes after treatment of cycloheximide, an inhibitor of protein biosynthesis (Fig. 4C). Therefore, under stress conditions, the keratin network might help to maintain the protein level of NF-κB, and NF-κB might play a role in the delayed apoptosis in K18-D238/397E hepatocytes. For example, we have previously shown binding between NF-kB and K8/K18 by coimmunoprecipitation (Lee et al., 2013), although the detailed molecular mechanism of their association remains to be determined.

A previous study demonstrated that the K18-D238/397E mutation in transgenic mice containing endogenous mouse K18-WT predisposed to hepatocyte necrosis during anti-FasAb-induced apoptotic liver injury (Weerasinghe et al., 2014). We compared the effect of K18-D238/397E mutation in transgenic mice without or with endogenous mouse K18-WT during hepatocyte necrosis. Primary hepatocytes from transgenic mice were prepared and treated with anti-FasAb and then the necrosis marker, high-mobility group box 1 protein (HMGB1) (Štros, 2010), was analyzed in hepatocyte culture medium. Under the tested condition, HMGB1 was not detected in the culture medium of K18-D238/397E hepatocytes from mice with an mK18-null background (K18-D238/397E;mK18-null mice), whereas increased levels of HMGB1was observed in the medium of control hepatocytes (Fig. S2). Consistent with the resistance of K18-D238/397E;mK18-null mice to liver injury in vivo (Fig. 2), this result indicated that K18-D238/397E mutation in mK18-null mice protects hepatocytes from necrosis during anti-FasAb-induced apoptotic liver injury. However, we could not observe a dramatic difference of HMGB1 levels in control hepatocytes and K18-D238/397E hepatocytes from transgenic mice containing endogenous mouse K18-WT, although HMGB1 levels in the mutant hepatocyte culture medium were slightly increased (Fig. S2).

Since K18-D238/397E hepatocytes treated with anti-FasAb show a relatively resistant keratin network (Fig. 3D and Fig. S1), we speculated that the increased protein stability of K18-D238/397E is related to enhanced hepatocyte integrity that protect the cells under stress. We, therefore, compared the stability of K18-D238/397E and K18-WT expressed in BHK cells after treating cells with cycloheximide (Fig. 5A). The stability of K18-D238/397E was increased up to 48 h after cycloheximide treatment, whereas K18-WT levels were reduced at 24 h concomitant with increased levels of the apoptotic K18 fragment (29 kD) (Fig. 5A) in the condition when the cell viability was greater than 90% (Fig. 5B). Consistent with this result, we observed the prolonged stability of K18-D238/397E protein in transfected cells after treatment with anisomycin, another protein synthesis inhibitor and apoptosis inducer (Fig. 5C). We also performed protein stability studies by using isolated primary hepatocytes after liver perfusion. Increased levels of K18 fragment (29 kD) coupled with decreased levels of intact K18 were detected in K18-WT hepatocytes but not in the mutant hepatocytes after cycloheximide treatment (Fig. 4C). Similar findings were made in livers of transgenic mice after administration of anti-FasAb (Fig. 5D and Fig. S3). Taken together, our findings show that the K18-D238/397E mutation enhances the protein stability of K18 (Fig. 5) and resistance to keratin network disruption under stress (Fig. 3D and Fig. S1), thereby providing hepatocyte integrity, leading to increased survival (Fig. 2).

Fig. 5.

K18-D238/397E protein has the enhanced protein stability. (A-C) BHK cells were transfected with K8/K18-WT or K8-WT/K18-D237/396E and incubated in (A,B) the presence or absence of cycloheximide (CHX; 100 μg/ml) or (C) anisomycin (AN; 10 μg/ml) for the indicated times. Total lysates were blotted for human K8 (hK8), human K18 (hK18) or actin. The graphs show the levels of intact K18 relative to those of actin. Data represent the mean±s.e.m. from three independent experiments. In B, cell viability after CHX treatment was determined using the Trypan Blue exclusion method and data represent the mean±s.e.m. from six independent experiments. (D) Total liver lysates as described in panel A in Fig. 4 were blotted using antibodies specifically recognizing human K18 (hK18) or mouse K8 (mK8). Levels of intact human K18-WT in hK18WT;mK18-null mice were markedly reduced after administration of anti-FasAb, whereas levels of intact human K18-D238/397E remained relatively stable. Note that levels of mouse K8 were almost similar in all tested total liver lysates.

Fig. 5.

K18-D238/397E protein has the enhanced protein stability. (A-C) BHK cells were transfected with K8/K18-WT or K8-WT/K18-D237/396E and incubated in (A,B) the presence or absence of cycloheximide (CHX; 100 μg/ml) or (C) anisomycin (AN; 10 μg/ml) for the indicated times. Total lysates were blotted for human K8 (hK8), human K18 (hK18) or actin. The graphs show the levels of intact K18 relative to those of actin. Data represent the mean±s.e.m. from three independent experiments. In B, cell viability after CHX treatment was determined using the Trypan Blue exclusion method and data represent the mean±s.e.m. from six independent experiments. (D) Total liver lysates as described in panel A in Fig. 4 were blotted using antibodies specifically recognizing human K18 (hK18) or mouse K8 (mK8). Levels of intact human K18-WT in hK18WT;mK18-null mice were markedly reduced after administration of anti-FasAb, whereas levels of intact human K18-D238/397E remained relatively stable. Note that levels of mouse K8 were almost similar in all tested total liver lysates.

The conserved caspase cleavage site in intermediate filaments

Cytoskeletal filaments, such as microtubules, intermediate filaments and microfilaments, provide structural scaffolding to cells in order to maintain their structure and cellular morphology (Goldman, 1971; Jordan and Wilson, 1998; Kawauchi and Hoshino, 2008; Ku et al., 1999; Pollard and Cooper, 2009; Rao and Li, 2004). In apoptotic cells, dramatic morphological changes occur, such as membrane blebbing, cell shrinkage and fragmentation of the nucleus and cell (Earnshaw, 1995; MacFarlane et al., 2000), with caspases playing crucial roles in these morphological changes (Cohen, 1997). Since cytoskeletal proteins, such as actin, actin-binding proteins, tubulin, tubulin-associated proteins and various intermediate-filament proteins, are cleaved by caspases in the presence of apoptotic signals and are also involved in cell morphology, proteolysis of these cytoskeletal proteins is likely to be related to apoptotic morphological changes in response to apoptosis (Taylor et al., 2008).

Intermediate filaments share a highly conserved caspase consensus sequence/caspase cleavage site (Table 1). Prior studies have demonstrated that caspase cleavage-resistant forms of intermediate filaments have a protective role after induction of an apoptosis stimulation. The first identified caspase-cleaved intermediate filament is lamin A, which forms the nuclear lamina and plays a structural role in the nucleus (Davidson and Lammerding, 2014; Worman and Schirmer, 2015; Zhang et al., 2001). Caspase cleavage sites in lamins include VEID230 in lamin A and VEVD231 in lamin B (Lazebnik et al., 1995; Rao et al., 1996). Inhibition of lamin cleavage by caspases prevents nuclear envelope disruption, nuclear and DNA fragmentation, and chromatin condensation and delays apoptosis (Rao et al., 1996; Ruchaud et al., 2002; Taylor et al., 2008; Zhang et al., 2001). Vimentin in mesenchymal cells has two caspase cleavage sites, DSVD85 and IDVD259, that are substrates of caspases-3/7 and caspases-6/8, respectively (Belichenko et al., 2001; Byun et al., 2001). Caspase-dependent vimentin cleavage is involved in vimentin filament disruption and can induce apoptosis (Belichenko et al., 2001; Byun et al., 2001). Desmin, the muscle-specific intermediate filament protein, is cleaved at VEMD263 by caspase 6 in vitro and its fragmentation promotes apoptosis in myogenic cells treated with TNFα (Chen et al., 2003). However, caspase cleavage-resistant desmin mutation (D236E) inhibits the desmin fragmentation and reduces the TNFα-induced apoptosis as compared with desmin WT in cell culture (Chen et al., 2003). These results were confirmed in corresponding transgenic mouse models. For example, comparison of cardiomyocyte apoptosis in mice that express endogenous WT desmin and mice that overexpress desmin D263E in a desmin-null background has shown that expression of D263E desmin delays TNFα-induced cardiomyocyte apoptosis and protects them from heart failure (Panagopoulou et al., 2008).

Table 1.

Effect of caspase cleavage-resistant filament during apoptosis

Effect of caspase cleavage-resistant filament during apoptosis
Effect of caspase cleavage-resistant filament during apoptosis

Fragmentation and disease relevance of keratin 18

K18, with its partner K8, makes up the intermediate filament cytoskeleton in adult hepatocytes, has two caspase cleavage sites, VEVD238 in the rod-domain and DALD397 in the C-terminal domain. K18 fragmentation occurs during apoptosis (Caulín et al., 1997; Ku et al., 1997; Leers et al., 1999), and fragmented K18 reorganizes into keratin aggregates that is released into the cell culture medium (Schutte et al., 2004). Indeed, caspase-cleaved K18 fragments are released into the blood of patients with liver diseases (Kramer et al., 2004; Yilmaz, 2009). The level of K18 fragments in blood is correlated with hepatic apoptosis, and the apoptotic K18 fragment is a useful biomarker to determine the degree of liver damage (Linder et al., 2010; Tsutsui et al., 2010).

Here, we studied the significance of K18 apoptotic cleavage under several stresses in transgenic mice that express the caspase cleavage-resistant K18 mutant K18-D238/397E in a mouse K18-null background. The phenotype of these mice clearly shows that expression of caspase cleavage-resistant K18 attenuates hepatic apoptosis and enhances the survival of the mice following liver damage induced by anti-FasAb or MLR, and indicates that an intact keratin network is crucial in order to protect cells from apoptotic stress. Our results agree with previous studies of cleavage-resistant lamins, and cleavage-resistant vimentin and desmin in cultured cells, and with a study of cleavage-resistant desmin in transgenic mice (Table 1). However, Weerasinghe et al.−by using K18-D238/397E-overexpressing transgenic mice that also express endogenous WT mouse K18 − have shown enhanced susceptibility to anti-FasAb-induced injury together with predisposition to hepatocyte necrosis (Weerasinghe et al., 2014). In our study, we also observed that the mutant transgenic mice that express endogenous mouse K18 show a slightly enhanced lethality in response to anti-FasAb-mediated liver damage as compared with control mice, but the difference was not statistically significant (Fig. 1). The phenotypic differences between the two mouse models (i.e. the heterozygous versus the homozygous effect) shown in Figs 1 and 2 are probably due to the endogenous mouse K18 in the heterozygous mutant transgenic mice attenuating the effect of the mutation. Our results clearly show enhanced protein stability of the mutant K18 compared with WT − both in vitro (cultured cells) and in vivo (transgenic livers) (Fig. 5) − that allows protection of the keratin network from liver damage (Fig. 3D). Indeed, the significance of an intact keratin network has initially been demonstrated in studies that used a mutated form of skin keratin 14 (K14-R125C) − that caused keratin filament disruption and, consequently, skin disease (Coulombe et al., 1991, 2009). Transgenic mice with the equivalent mutation of K18 (K18-R90C) have disrupted filaments and increased susceptibility to liver injury (Ku et al., 1995; Omary et al., 2009). Both cases highlight the significance of an intact keratin network in protecting cells from damage. Our finding provides evidence that the enhanced protein stability of caspase cleavage-resistant keratin 18 contributes towards maintaining the keratin network under stress.

Reagents

Antibodies against keratins have been previously described (Ku et al., 2004). We also used antibodies against: Fas (anti-FasAb: BD Biosciences, San Jose, CA), phosphorylated or nonphosphorylated-protein kinases or transcription factors (Cell Signaling Technology, Beverly, MA) such as Akt, p42MAPK and p44MAPK (also known as MAPK1 and MAPK3, respectively), p90RSK (also known as RPS6KA1), p38MAPK (also known as MAPK14), JNK1 (also known as MAPK8), and JNK2 (also known as MAPK9), against cleaved caspase 3, 7 or 8 and PARP (Cell Signaling Technology, Beverly, MA), Hsc70 (Santa Cruz Biotechnology, Santa Cruz, CA), FADD (Upstate Biotechnology/Millipore, Billerica, MA), HMGB1 (Abcam, Cambridge, MA), and actin (Lab Vision/Thermo Scientific, Fremont, CA) (see Table S1 for details on antibodies). Other reagents included: MLR (used at 30 μg/kg body weight; Alexis Corporation, San Diego, CA), anisomycin (10 μg/ml; CalBiochem, La Jolla, CA) and cycloheximide (100 μg/ml; Sigma-Aldrich, St Louis, MO).

Generation of K18-WT or D238/397E transgenic mice in a K18-null background

Heterozygous transgenic mice expressing human K18-WT or human K18-D238/397E in an FVB/N background (Weerasinghe et al., 2014) were crossbred with K18-null mice. By using PCR, the genome of mouse progenies was screened for the presence of human K18 gene and the absence of mouse K18 as described (Ku et al., 1998; Magin et al., 1998). K18-null mice were kindly provided by Dr Thomas Margin (University of Leipzig, Leipzig, Germany) (Magin et al., 1998) and, as they originally had been of a mixed strain background, were backcrossed ten generations to an FVB/N background. All animal experiments were performed in accordance with the Korean Food and Drug Administration (KFDA) guidelines. Protocols were approved by the Institutional Animal Care and Use Committee (IACUC) of the Yonsei Laboratory Animal Research Center (YLARC).

Toxin administration and liver tissue preparation

Mice were starved overnight and intraperitoneally injected with anti-FasAb (0.15 mg/kg body weight), or with MLR (30 μg/kg body weight) as previously described (Ku et al., 2004). The mice were assessed for three days for mortality experiments. Alternatively, mice were killed at 6 or 8 h after injection with anti-FasAb, and their livers were isolated and used for biochemical analysis, histological analysis or immunostaining. For biochemical analysis, livers were homogenized in ice-cold phosphate-buffered saline (PBS) containing 5 mM EDTA, sodium fluoride (NaF), sodium pyrophosphate (NaPP), a protease inhibitor (25 mg/ml aprotinin, 10 mM leupeptin, 10 mM pepstatin and 0.1 mM PMSF) and the phosphatase inhibitor okadaic acid. Total liver lysates and high-salt extracts were prepared by using total liver homogenates as described (Ku et al., 2004). For tissue staining, livers were either fixed with 10% formalin, embedded in paraffin and sectioned for hematoxylin-eosin staining, or embedded in optimum cutting temperature compound and sectioned for immunostaining.

Hepatocyte isolation

Primary hepatocytes from K18-WT or K18-D238/397E mice were isolated by the liver perfusion method as described previously (Ku and Omary, 2006). Isolated hepatocytes were cultured in HepatoZYME serum-free medium (Gibco BRL Life Technologies, Grand Island, NY) with 10% fetal calf serum for 24 h and then treated with anti-FasAb (0.5 μg/ml or 1.5 μg/ml for 6−36 h) or cycloheximide (100 μg/ml). Cultured hepatocytes were used for immunostaining or preparations of total cell lysates were used for immunoblotting.

Cell culture and transfection

Baby hamster kidney (BHK)-21 cells (American Type Culture Collection, Rockville, MD) were cultured in medium as recommended by the supplier. Transient transfection was performed by using Lipofectamine (Invitrogen, Carlsbad, CA) according to the manufacturer's instructions. Transfected cells were treated with anisomycin (10 μg/ml) or cycloheximide (100 μg/ml).

Immunoblot analysis

Total lysates and high-salt extract samples were analyzed by SDS-PAGE followed by Coomassie Blue staining or immunoblotting with antibodies as indicated. Proteins were then visualized using enhanced chemiluminescence.

Statistical analysis

Statistical comparisons were performed using Fisher's exact test in StatView software. P-values of ≤0.05 were considered significant.

We thank Dr Thomas Magin for providing us with the K18-null mice and Dr Bishr Omary (University of Michigan, Ann Arbor, USA) for providing us with the heterozygous K18-WT and K18-D238/397E transgenic mice.

Author contributions

Conceptualization: H.Y., N.K.; Methodology: H.Y., N.K.; Validation: H.Y., N.K.; Formal analysis: H.Y., S.Y., J.H.; Investigation: H.Y., S.Y., J.H.; Writing - original draft: H.Y., N.K.; Writing - review & editing: N.K.; Supervision: N.K.

Funding

This work was supported by the Korean Ministry of Education, Science and Technology (grant number 2016R1A2B4012808 to N.-O.K.).

Belichenko
,
I.
,
Morishima
,
N.
and
Separovic
,
D.
(
2001
).
Caspase-resistant vimentin suppresses apoptosis after photodynamic treatment with a silicon phthalocyanine in Jurkat cells
.
Arch. Biochem. Biophys.
390
,
57
-
63
.
Byun
,
Y.
,
Chen
,
F.
,
Chang
,
R.
,
Trivedi
,
M.
,
Green
,
K. J.
and
Cryns
,
V. L.
(
2001
).
Caspase cleavage of vimentin disrupts intermediate filaments and promotes apoptosis
.
Cell Death Differ.
8
,
443
-
450
.
Caulín
,
C.
,
Salvesen
,
G. S.
and
Oshima
,
R. G.
(
1997
).
Caspase cleavage of keratin 18 and reorganization of intermediate filaments during epithelial cell apoptosis
.
J. Cell Biol.
138
,
1379
-
1394
.
Chen
,
F.
,
Chang
,
R.
,
Trivedi
,
M.
,
Capetanaki
,
Y.
and
Cryns
,
V. L.
(
2003
).
Caspase proteolysis of desmin produces a dominant-negative inhibitor of intermediate filaments and promotes apoptosis
.
J. Biol. Chem.
278
,
6848
-
6853
.
Cohen
,
G. M.
(
1997
).
Caspases: the executioners of apoptosis
.
Biochem. J.
326
,
1
-
16
.
Coulombe
,
P. A.
,
Hutton
,
M. E.
,
Letal
,
A.
,
Hebert
,
A.
,
Paller
,
A. S.
and
Fuchs
,
E.
(
1991
).
Point mutations in human keratin 14 genes of epidermolysis bullosa simplex patients: genetic and functional analyses
.
Cell
66
,
1301
-
1311
.
Coulombe
,
P. A.
,
Kerns
,
M. L.
and
Fuchs
,
E.
(
2009
).
Epidermolysis bullosa simplex: a paradigm for disorders of tissue fragility
.
J. Clin. Invest.
119
,
1784
-
1793
.
Davidson
,
P. M.
and
Lammerding
,
J.
(
2014
).
Broken nuclei–lamins, nuclear mechanics, and disease
.
Trends Cell Biol.
24
,
247
-
256
.
Earnshaw
,
W. C.
(
1995
).
Nuclear changes in apoptosis
.
Curr. Opin. Cell Biol.
7
,
337
-
343
.
Eriksson
,
J. E.
,
Dechat
,
T.
,
Grin
,
B.
,
Helfand
,
B.
,
Mendez
,
M.
,
Pallari
,
H.-M.
and
Goldman
,
R. D.
(
2009
).
Introducing intermediate filaments: from discovery to disease
.
J. Clin. Invest.
119
,
1763
-
1771
.
Goldman
,
R. D.
(
1971
).
The role of three cytoplasmic fibers in BHK-21 cell motility. I. Microtubules and the effects of colchicine
.
J. Cell Biol.
51
,
752
-
762
.
Herrmann
,
H.
,
Strelkov
,
S. V.
,
Burkhard
,
P.
and
Aebi
,
U.
(
2009
).
Intermediate filaments: primary determinants of cell architecture and plasticity
.
J. Clin. Invest.
119
,
1772
-
1783
.
Jochimsen
,
E. M.
,
Carmichael
,
W. W.
,
An
,
J. S.
,
Cardo
,
D. M.
,
Cookson
,
S. T.
,
Holmes
,
C. E. M.
,
Antunes
,
M. B.
,
de Melo Filho
,
D. A.
,
Lyra
,
T. M.
,
Barreto
,
V. S. T.
, et al. 
(
1998
).
Liver failure and death after exposure to microcystins at a hemodialysis center in Brazil
.
N. Engl. J. Med.
338
,
873
-
878
.
Jordan
,
M. A.
and
Wilson
,
L.
(
1998
).
Microtubules and actin filaments: dynamic targets for cancer chemotherapy
.
Curr. Opin. Cell Biol.
10
,
123
-
130
.
Kawauchi
,
T.
and
Hoshino
,
M.
(
2008
).
Molecular pathways regulating cytoskeletal organization and morphological changes in migrating neurons
.
Dev. Neurosci.
30
,
36
-
46
.
Kramer
,
G.
,
Erdal
,
H.
,
Mertens
,
H. J.
,
Nap
,
M.
,
Mauermann
,
J.
,
Steiner
,
G.
,
Marberger
,
M.
,
Biven
,
K.
,
Shoshan
,
M. C.
and
Linder
,
S.
(
2004
).
Differentiation between cell death modes using measurements of different soluble forms of extracellular cytokeratin 18
.
Cancer Res.
64
,
1751
-
1756
.
Ku
,
N.-O.
and
Omary
,
M. B.
(
2001
).
Effect of mutation and phosphorylation of type I keratins on their caspase-mediated degradation
.
J. Biol. Chem.
276
,
26792
-
26798
.
Ku
,
N.-O.
and
Omary
,
M. B.
(
2006
).
A disease- and phosphorylation-related nonmechanical function for keratin 8
.
J. Cell Biol.
174
,
115
-
125
.
Ku
,
N. O.
,
Michie
,
S.
,
Oshima
,
R. G.
and
Omary
,
M. B.
(
1995
).
Chronic hepatitis, hepatocyte fragility, and increased soluble phosphoglycokeratins in transgenic mice expressing a keratin 18 conserved arginine mutant
.
J. Cell Biol.
131
,
1303
-
1314
.
Ku
,
N.-O.
,
Liao
,
J.
and
Omary
,
M. B.
(
1997
).
Apoptosis generates stable fragments of human type I keratins
.
J. Biol. Chem.
272
,
33197
-
33203
.
Ku
,
N.-O.
,
Michie
,
S. A.
,
Soetikno
,
R. M.
,
Resurreccion
,
E. Z.
,
Broome
,
R. L.
and
Omary
,
M. B.
(
1998
).
Mutation of a major keratin phosphorylation site predisposes to hepatotoxic injury in transgenic mice
.
J. Cell Biol.
143
,
2023
-
2032
.
Ku
,
N. O.
,
Zhou
,
X.
,
Toivola
,
D. M.
and
Omary
,
M. B.
(
1999
).
The cytoskeleton of digestive epithelia in health and disease
.
Am. J. Physiol.
277
,
G1108
-
G1137
.
Ku
,
N.-O.
,
Toivola
,
D. M.
,
Zhou
,
Q.
,
Tao
,
G.-Z.
,
Zhong
,
B.
and
Omary
,
M. B.
(
2004
).
Studying simple epithelial keratins in cells and tissues
.
Methods Cell Biol.
78
,
489
-
517
.
Ku
,
N.-O.
,
Strnad
,
P.
,
Bantel
,
H.
and
Omary
,
M. B.
(
2016
).
Keratins: biomarkers and modulators of apoptotic and necrotic cell death in the liver
.
Hepatology
64
,
966
-
976
.
Lazebnik
,
Y. A.
,
Takahashi
,
A.
,
Moir
,
R. D.
,
Goldman
,
R. D.
,
Poirier
,
G. G.
,
Kaufmann
,
S. H.
and
Earnshaw
,
W. C.
(
1995
).
Studies of the lamin proteinase reveal multiple parallel biochemical pathways during apoptotic execution
.
Proc. Natl. Acad. Sci. USA
92
,
9042
-
9046
.
Lee
,
J.
,
Jang
,
K.-H.
,
Kim
,
H.
,
Lim
,
Y.
,
Kim
,
S.
,
Yoon
,
H.-N.
,
Chung
,
I. K.
,
Roth
,
J.
and
Ku
,
N.-O.
(
2013
).
Predisposition to apoptosis in keratin 8-null liver is related to inactivation of NF-kappaB and SAPKs but not decreased c-Flip
.
Biol. Open
2
,
695
-
702
.
Leers
,
M. P. G.
,
Kolgen
,
W.
,
Bjorklund
,
V.
,
Bergman
,
T.
,
Tribbick
,
G.
,
Persson
,
B.
,
Bjorklund
,
P.
,
Ramaekers
,
F. C. S.
,
Bjorklund
,
B.
,
Nap
,
M.
, et al. 
(
1999
).
Immunocytochemical detection and mapping of a cytokeratin 18 neo-epitope exposed during early apoptosis
.
J. Pathol.
187
,
567
-
572
.
Linder
,
S.
,
Olofsson
,
M. H.
,
Herrmann
,
R.
and
Ulukaya
,
E.
(
2010
).
Utilization of cytokeratin-based biomarkers for pharmacodynamic studies
.
Expert Rev. Mol. Diagn.
10
,
353
-
359
.
MacFarlane
,
M.
,
Merrison
,
W.
,
Dinsdale
,
D.
and
Cohen
,
G. M.
(
2000
).
Active caspases and cleaved cytokeratins are sequestered into cytoplasmic inclusions in TRAIL-induced apoptosis
.
J. Cell Biol.
148
,
1239
-
1254
.
Magin
,
T. M.
,
Schröder
,
R.
,
Leitgeb
,
S.
,
Wanninger
,
F.
,
Zatloukal
,
K.
,
Grund
,
C.
and
Melton
,
D. W.
(
1998
).
Lessons from keratin 18 knockout mice: formation of novel keratin filaments, secondary loss of keratin 7 and accumulation of liver-specific keratin 8-positive aggregates
.
J. Cell Biol.
140
,
1441
-
1451
.
Ogasawara
,
J.
,
Watanabe-Fukunaga
,
R.
,
Adachi
,
M.
,
Matsuzawa
,
A.
,
Kasugai
,
T.
,
Kitamura
,
Y.
,
Itoh
,
N.
,
Suda
,
T.
and
Nagata
,
S.
(
1993
).
Lethal effect of the anti-Fas antibody in mice
.
Nature
364
,
806
-
809
.
Omary
,
M. B.
,
Ku
,
N.-O.
,
Strnad
,
P.
and
Hanada
,
S.
(
2009
).
Toward unraveling the complexity of simple epithelial keratins in human disease
.
J. Clin. Invest.
119
,
1794
-
1805
.
Panagopoulou
,
P.
,
Davos
,
C. H.
,
Milner
,
D. J.
,
Varela
,
E.
,
Cameron
,
J. A.
,
Mann
,
D. L.
and
Capetanaki
,
Y.
(
2008
).
Desmin mediates TNF-alpha-induced aggregate formation and intercalated disk reorganization in heart failure
.
J. Cell Biol.
181
,
761
-
775
.
Pollard
,
T. D.
and
Cooper
,
J. A.
(
2009
).
Actin, a central player in cell shape and movement
.
Science
326
,
1208
-
1212
.
Rao
,
J.
and
Li
,
N.
(
2004
).
Microfilament actin remodeling as a potential target for cancer drug development
.
Curr. Cancer Drug Targets
4
,
345
-
354
.
Rao
,
L.
,
Perez
,
D.
and
White
,
E.
(
1996
).
Lamin proteolysis facilitates nuclear events during apoptosis
.
J. Cell Biol.
135
,
1441
-
1455
.
Riedl
,
S. J.
and
Shi
,
Y.
(
2004
).
Molecular mechanisms of caspase regulation during apoptosis
.
Nat. Rev. Mol. Cell Biol.
5
,
897
-
907
.
Ruchaud
,
S.
,
Korfali
,
N.
,
Villa
,
P.
,
Kottke
,
T. J.
,
Dingwall
,
C.
,
Kaufmann
,
S. H.
and
Earnshaw
,
W. C.
(
2002
).
Caspase-6 gene disruption reveals a requirement for lamin A cleavage in apoptotic chromatin condensation
.
EMBO J.
21
,
1967
-
1977
.
Schutte
,
B.
,
Henfling
,
M.
,
Kölgen
,
W.
,
Bouman
,
M.
,
Meex
,
S.
,
Leers
,
M. P. G.
,
Nap
,
M.
,
Björklund
,
V.
,
Björklund
,
P.
,
Bjorklund
,
B.
, et al. 
(
2004
).
Keratin 8/18 breakdown and reorganization during apoptosis
.
Exp. Cell Res.
297
,
11
-
26
.
Snider
,
N. T.
and
Omary
,
M. B.
(
2014
).
Post-translational modifications of intermediate filament proteins: mechanisms and functions
.
Nat. Rev. Mol. Cell Biol
15
,
163
-
177
.
Štros
,
M.
(
2010
).
HMGB proteins: interactions with DNA and chromatin
.
Biochim. Biophys. Acta
1799
,
101
-
113
.
Taylor
,
R. C.
,
Cullen
,
S. P.
and
Martin
,
S. J.
(
2008
).
Apoptosis: controlled demolition at the cellular level
.
Nat. Rev. Mol. Cell Biol.
9
,
231
-
241
.
Tsutsui
,
M.
,
Tanaka
,
N.
,
Kawakubo
,
M.
,
Sheena
,
Y.
,
Horiuchi
,
A.
,
Komatsu
,
M.
,
Nagaya
,
T.
,
Joshita
,
S.
,
Umemura
,
T.
,
Ichijo
,
T.
, et al. 
(
2010
).
Serum fragmented cytokeratin 18 levels reflect the histologic activity score of nonalcoholic fatty liver disease more accurately than serum alanine aminotransferase levels
.
J. Clin. Gastroenterol.
44
,
440
-
447
.
Weerasinghe
,
S. V. W.
,
Ku
,
N.-O.
,
Altshuler
,
P. J.
,
Kwan
,
R.
and
Omary
,
M. B.
(
2014
).
Mutation of caspase-digestion sites in keratin 18 interferes with filament reorganization, and predisposes to hepatocyte necrosis and loss of membrane integrity
.
J. Cell Sci.
127
,
1464
-
1475
.
Worman
,
H. J.
and
Schirmer
,
E. C.
(
2015
).
Nuclear membrane diversity: underlying tissue-specific pathologies in disease?
Curr. Opin. Cell Biol.
34
,
101
-
112
.
Yilmaz
,
Y.
(
2009
).
Systematic review: caspase-cleaved fragments of cytokeratin 18 - the promises and challenges of a biomarker for chronic liver disease
.
Aliment. Pharmacol. Ther.
30
,
1103
-
1109
.
Zhang
,
D.
,
Beresford
,
P. J.
,
Greenberg
,
A. H.
and
Lieberman
,
J.
(
2001
).
Granzymes A and B directly cleave lamins and disrupt the nuclear lamina during granule-mediated cytolysis
.
Proc. Natl. Acad. Sci. USA
98
,
5746
-
5751
.

Competing interests

The authors declare no competing or financial interests.

Supplementary information