Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Cycloheximide Can Induce Bax/Bak Dependent Myeloid Cell Death Independently of Multiple BH3-Only Proteins

  • Katharine J. Goodall ,

    katharine.goodall@monash.edu

    Affiliation Department of Biochemistry, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, Australia

  • Megan L. Finch-Edmondson,

    Affiliation School of Chemistry and Biochemistry, University of Western Australia, Crawley, WA, Australia

  • Joanne van Vuuren,

    Affiliation Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, the University of Western Australia, Crawley, WA, Australia

  • George C. Yeoh,

    Affiliations School of Chemistry and Biochemistry, University of Western Australia, Crawley, WA, Australia, Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands and Centre for Medical Research, the University of Western Australia, Crawley, WA, Australia

  • Ian E. Gentle,

    Affiliations Department of Biochemistry, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, Australia, Institute for Medical Microbiology and Hygiene, Hermann-Herder-Str. 11 79104 Freiburg Germany

  • James E. Vince,

    Affiliations Department of Biochemistry, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, Australia, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia, Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia

  • Paul G. Ekert,

    Affiliations Department of Biochemistry, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, Australia, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia, Murdoch Childrens Research Institute, Royal Children's Hospital, Parkville, VIC, Australia

  • David L. Vaux,

    Affiliations Department of Biochemistry, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, Australia, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia, Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia

  • Bernard A. Callus

    Affiliations Department of Biochemistry, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, Australia, School of Chemistry and Biochemistry, University of Western Australia, Crawley, WA, Australia, School of Health Sciences, The University of Notre Dame Australia, Fremantle, WA, Australia

Abstract

Apoptosis mediated by Bax or Bak is usually thought to be triggered by BH3-only members of the Bcl-2 protein family. BH3-only proteins can directly bind to and activate Bax or Bak, or indirectly activate them by binding to anti-apoptotic Bcl-2 family members, thereby relieving their inhibition of Bax and Bak. Here we describe a third way of activation of Bax/Bak dependent apoptosis that does not require triggering by multiple BH3-only proteins. In factor dependent myeloid (FDM) cell lines, cycloheximide induced apoptosis by a Bax/Bak dependent mechanism, because Bax-/-Bak-/- lines were profoundly resistant, whereas FDM lines lacking one or more genes for BH3-only proteins remained highly sensitive. Addition of cycloheximide led to the rapid loss of Mcl-1 but did not affect the expression of other Bcl-2 family proteins. In support of these findings, similar results were observed by treating FDM cells with the CDK inhibitor, roscovitine. Roscovitine reduced Mcl-1 abundance and caused Bax/Bak dependent cell death, yet FDM lines lacking one or more genes for BH3-only proteins remained highly sensitive. Therefore Bax/Bak dependent apoptosis can be regulated by the abundance of anti-apoptotic Bcl-2 family members such as Mcl-1, independently of several known BH3-only proteins.

Introduction

The role of Bcl-2 as an inhibitor of cell death was first established in FDC-P1 cells, an IL-3 dependent mouse myeloid cell line [1]. These cells undergo apoptosis when growth factor is removed, but when growth factor was removed from cells over-expressing Bcl-2, they arrested, but did not die.

Similar factor dependent myeloid (FDM) cell lines have been generated by infecting murine bone marrow or foetal liver cells with retroviruses expressing HoxB8, and culturing in IL-3 [25]. FDM lines lacking genes for pro-apoptotic members of the Bcl-2 family, such as the multi-domain proteins (Bax and Bak), or a number of BH3-only proteins, have been generated in a similar way by using bone marrow or foetal liver from gene deleted mice. In this way we have obtained IL-3 dependent myeloid lines lacking genes for Bax or Bak, Bax and Bak, Blk (Bik), Puma, Noxa, Bim, Bad, Bim, Bmf and Hrk as well as lines lacking both Bim and Bad, both Bim and Bid and both Puma and Noxa. All of these cell lines remain dependent on cytokines for growth and proliferation.

Cycloheximide (CHX) is an inhibitor of protein synthesis [6]. Many cell types rapidly undergo apoptosis when exposed to CHX. In FDC-P1 cells, CHX-induced apoptosis is mediated by Bax and/or Bak because it can be inhibited by over-expression of Bcl-2 [7].

Bax/Bak dependent apoptosis is widely believed to be triggered by BH3-only proteins and that they have an essential and obligatory role in the activation of Bax and/or Bak [8, 9]. The BH3-only members such as Bik, Bid, Bim, Bad, Puma, Noxa, Bmf and Hrk fall into two classes. The “direct activators”, such as Bid, Bim and Puma, can bind directly to Bak or Bax to activate them. Members of the other class, the “indirect activators”, which includes Bad, Bik, Bmf, Hrk and Noxa, act by binding to anti-apoptotic Bcl-2 family members (namely Mcl-1, Bcl-2, Bcl-x, A1 and Bcl-w) and thereby prevent them from inhibiting Bax or Bax [10].

To determine which BH3-only protein(s) were responsible for apoptosis of FDM cells in response to cycloheximide, we compared the sensitivity of cell lines mutant for various pro-apoptotic Bcl-2 family members.

We were surprised to find that none of the lines lacking genes for individual BH3-only proteins were resistant to CHX induced apoptosis, and furthermore, lines lacking both Bim and Bid, and with undetectable levels of Puma [4], still underwent apoptosis in response to CHX.

Collectively these results indicate that CHX does not induce FDM cell death by activation of BH3-only proteins, but that activation of Bax/Bak and apoptosis in this case is caused by a reduction in the abundance of Mcl-1. Furthermore, they suggest that loss of one or more pro-survival proteins can be sufficient to permit activation of Bax/Bak, and that in some circumstances Bax and Bak can be activated in the absence of BH3-only proteins involvement.

Results

Initially, a dose-response experiment was performed to determine the concentration of cycloheximide (CHX) that caused FDM cells to die. CHX induced a dose-dependent decrease in viability of wild-type (WT) FDM cells for concentrations above 1 μg/ml with greater than 90% of cells killed at 20 μg/ml by 24 h (Fig 1A). CHX induced cell death by this time point was dependent on the expression of Bax or Bak because bax/bak deficient FDM cells derived from bax/bak double knockout (DKO) mice were profoundly resistant to CHX treatment (Fig 1A). This resistance was confirmed by treating bax/bak DKO cells for 96 hours with CHX, at which the majority of the cells were still viable, in contrast to the rapid cell death of the WT cells (Fig 1B) From these experiments a concentration of 20 μg/ml CHX was chosen and was used in all subsequent experiments.

thumbnail
Fig 1. CHX induces a time and dose-dependent Bax/Bak-dependent death in FDM cells.

A) WT and bax/bak DKO cells were treated with CHX at the concentrations indicated for 24 h before determining cell viability by PI exclusion and flow cytometry. bax/bak (B) and WT (C) cells were treated with 20 μg/ml CHX for the times indicated before cell viability was determined. D) WT cells were incubated with 20 μg/ml CHX for the times indicated before being washed and resuspended in fresh media without CHX. Cultures were continued before cell viability was determined at 24 h. Data presented are expressed as mean ± SEM from 3 independent experiments.

https://doi.org/10.1371/journal.pone.0164003.g001

Next, WT cells were treated with CHX for varying times to determine how rapidly it induced cell death. In this experiment, most of the cells had died by 16 h (Fig 1C). To determine whether the effects of CHX treatment were reversible, WT FDM cells were treated for between 2 and 24 h with 20 μg/ml CHX, and then washed and resuspended in CHX-free media, and their viability was measured at 24 h. As seen in Fig 1D, incubating cells for less than 2–6 h with CHX had only modest effects on cell viability measured at 24 h. However, incubating for more than 8 h caused over half the cells to die by 24 h. Collectively, these experiments show that cells treated with CHX do not die immediately, but when exposed for greater than 8–24 h become irreversibly committed to die by a Bax/Bak dependent process.

In order to determine the requirement for BH3-only proteins in the activation of Bax/Bak in this system, we examined the ability of CHX to induce death of FDM cell lines derived from mice in which the genes encoding various BH3-only proteins were deleted. Consistent with the previous results, two independent WT clones of FDM cells were highly sensitive to CHX, with a significant loss in viability observed at 24 h post-treatment (Fig 2A). Again bax/bak DKO cells were highly resistant to CHX treatment. Next, FDM cells bearing gene deletions for a single BH3-only protein were examined for their sensitivity to CHX treatment. Remarkably, none of the BH3-only single knockout (SKO) cell lines examined exhibited any resistance towards CHX (Fig 2B). One of the bid knockout cell lines, bid 1, showed slightly increased resistance to CHX-induced toxicity yet the other bid-/- clone, bid 2, did not exhibit the same insensitivity. This suggests that the slight resistance in the bid 1 line is due to clonal variation.

thumbnail
Fig 2. CHX induces death in FDM cells independently of several known BH3-only proteins.

Viability of (A) WT and bax/bak DKO, (B) single BH3-only knockout and (C) BH3-only DKO FDM cell lines were determined after 24 h of treatment with 20 μg/ml CHX. (D) WT, bax/bak and BH3-only knockout cells were treated with 20 μg/ml CHX for the times indicated before cell viability was determined. Data shown is expressed as mean ± SEM from 3 independent experiments. The numbers after a cell line’s name refer to independent clonal lines of the same genotype.

https://doi.org/10.1371/journal.pone.0164003.g002

Next, clones of FDM DKO cells containing combinations of BH3-only protein gene deletions were examined for their sensitivity to CHX. Similar to the SKO results, none of the DKO cells lines examined exhibited any protection towards CHX-induced cell death (Fig 2C). It is also worth noting that despite the bid 1 cells exhibiting a slight resistance to CHX (Fig 2B), neither of the bid/bim DKO cell lines showed a similar resistance (Fig 2C), further suggesting that the slight resistance observed with bid 1 cells is due to clonal variation. To rule out the possibility that cell lines lacking BH3-only proteins might show resistance to CHX at earlier time points, we performed a time-course experiment using several knockout cell lines. Consistent with previous data, bax/bak DKO lines showed profound resistance to CHX (Fig 2D). In contrast, the bad, puma, bim and blk SKO lines as well as the bid/bim DKO cells lost viability at similar rates to that of WT cells following CHX addition. Although our entire panel of cell lines was not tested, these results suggest that the absence of one of more BH3-only proteins does not prevent or delay activation of Bax/Bak dependent cell death even at early times following CHX treatment.

The uptake of PI into dead or dying cells due to membrane damage is a relatively late event in the apoptotic process. Since the effects of CHX that resulted in cell death occurred between 8–16 h of treatment (Fig 1B), we looked for changes that might lead to activation of Bax or Bak within 8 h of CHX addition. Because the results in Fig 2 argued that none of the members of the BH3-only protein family examined were essential for activation of Bax or Bak, we looked to see whether addition of CHX might trigger apoptosis by causing a reduction in the levels of an anti-apoptotic Bcl-2 family member.

We determined the effect of CHX on the levels of the Bcl-2 pro-survival family members in both WT and bax/bak DKO cells (Fig 3). The advantage afforded by the bax/bak DKO cells is that changes in protein expression can be determined without confounding effects of cell death, such as caspase activation, which occurred in the WT cells. Levels of Bcl-2, Bcl-xL and Bcl-w proteins were not affected by CHX in either WT or bax/bak DKO cells within the 8 h time course, indicating these are stable proteins with long half-lives (Fig 3A). Quantification data is of levels of pro-survival proteins in Bax/Bak DKO cells after addition of CHX, only showing the larger, transient band of Mcl-1. A1 was not detected in these cells. Unlike the other pro-survival proteins, the abundance of Mcl-1 rapidly decreased in both WT and bax/bak DKO FDM lines. Mcl-1 exists as a doublet, with the smaller band being a more stable form in which the N-terminal tail has been truncated [11, 12]. However, the larger band has a shorter half-life, and in WT cells, this form of Mcl-1 fell to barely detectable levels within 2 h of CHX treatment. In bax/bak DKO cells, the abundance of Mcl-1 in untreated cells was much higher than in the WT cells, but the addition of CHX caused a rapid decrease in levels of the unstable form of Mcl-1 such that it was barely detectable within 4–8 h of treatment. These results suggest that loss of Mcl-1 may be responsible for Bax/Bak activation following CHX addition.

thumbnail
Fig 3. CHX induces the proteasomal loss of Mcl-1, but not other pro-survival Bcl-2 family proteins.

A) Lysates of WT and bax/bak DKO cells treated with 20 μg/ml CHX for 0–8 h were separated by SDS-PAGE, transferred to membranes and immunoblotted with antibodies to Mcl-1, Bcl-2, Bcl-xL and Bcl-w. 293T cells transiently expressing mouse Bcl-xL, and FDCP1 cells expressing Bcl-w, were used as positive controls for the Bcl-xL and Bcl-w antibodies. ß-actin was used a loading control. B) Densitometry analysis of western blots probed with pro-survival proteins, normalized to 0h timepoint. Mcl-1 densitometry refers only to the transient form of the protein, in bax/bak DKO cells. Results are representative of three independent experiments. C) WT and bax/bak DKO cells were pre-incubated with or without 20 μM MG132 before being treated with or without 20 μg/ml CHX as indicated. Cell lysates were immunoblotted for Mcl-1 and ß-actin as shown. Asterisks indicate cleavage products of Mcl-1 and cross-reactive species.

https://doi.org/10.1371/journal.pone.0164003.g003

To examine whether the loss of Mcl-1 in the presence of CHX was mediated by the proteasome, WT and bax/bak DKO cells were treated with CHX for 4 h with or without a 1 h pre-incubation with the proteasome inhibitor, MG132. Treatment of both cell lines with MG132 caused an increase in the levels of Mcl-1, indicating that its abundance in resting cells is limited by turnover within the proteasome (Fig 3C). Consistent with the previous results, addition of CHX alone led to the complete loss of the unstable form of Mcl-1 within 4 hours. Significantly, pre-treatment of cells with MG132 prevented the loss of Mcl-1, indicating that normal proteasomal turnover of Mcl-1 accounts for the loss of Mcl-1 following inhibition of protein synthesis with CHX. As MG132 prevented degradation of Mcl-1 following the addition of CHX, we then examined whether MG132 could prevent CHX-induced cell death in wild-type FDM cells. Unfortunately, treatment with low concentrations of MG132 were highly toxic to the FDM cells, thus this possibility could not be examined in these cells.

The loss of Mcl-1 following CHX treatment suggests that another means of reducing Mcl-1 might also initiate cell death in a BH3-independent manner, similar to CHX. Roscovitine, a CDK inhibitor, has been shown to inhibit Mcl-1 transcription [13]. Consistent with this, the treatment of FDM cells with roscovitine results in the rapid loss of Mcl-1 (Fig 4A). To investigate its effects on viability, cells were then treated with increasing concentrations of roscovitine, which induced profound cell death in a Bax/Bak dependent manner since bax/bak DKO cells were highly resistant to its effects (Fig 4B). Next we tested our panel of FDM knockout cell lines with roscovitine, and consistent with out CHX data, none of the BH3-only knockout cell lines showed any significant protection against roscovitine (Fig 4C). These results suggest that, like CHX-induced cell death, the BH3-only proteins are not involved in the initiation of roscovitine-induced FDM cell death but rather it is the loss of Mcl-1 that triggers activation of cell death.

thumbnail
Fig 4. Roscovitine reduces levels of Mcl-1 and induces death in FDM cells independently of BH3-only proteins.

A) Lysates of WT and baax/bak DKO FDM cells treated with roscovitine (30 μM) for the indicated times were separated by SDS-PAGE, transferred to membrane and immunoblotted with the Mcl-1. β-actin was used a loading control. B) WT cells were treated with increasing concentrations of roscovitine before cell viability was determined. C) WT, bax/bak and BH3-only knockout cells were treated with 30 μg/ml roscovitine for 24 h before cell viability was determined. Data shown is expressed as mean ± SEM from 3 independent experiments. The numbers after a cell line’s name refer to independent clonal lines of the same genotype.

https://doi.org/10.1371/journal.pone.0164003.g004

As with addition of CHX, withdrawal of IL-3 from the culture media of WT FDM cells causes a loss of viability over 48 h, whereas bax/bak DKO cells are completely resistant to this apoptotic stimulus (Fig 5A) [2]. In order to determine whether withdrawal of IL-3 also caused a reduction in Mcl-1, we examined its abundance in lysates of cells at 24 and 48 h after IL-3 withdrawal. Mcl-1 was not detected in WT cells 24 h after IL-3 removal (Fig 5B), but its absence might have been a consequence of cell death, or a cause, or both. In bax/bak DKO cells, Mcl-1 levels were reduced, but by a lesser extent, and it was still detectable even at 48 h following IL-3 withdrawal (Fig 5B).

thumbnail
Fig 5. IL-3 withdrawal reduces Mcl-1 abundance.

A) WT and bax/bak DKO cells were washed and incubated in media without IL-3. Cell viability was determined at 24 and 48 h after IL-3 removal. Data shown are from a single experiment. B) Cell lysates were prepared at 0, 24 and 48 h following IL-3 withdrawal and immunoblotted for Mcl-1 and ß-actin as shown.

https://doi.org/10.1371/journal.pone.0164003.g005

To confirm that lack of Mcl-1 was responsible for the death of CHX treated FDM cells, we tested whether over-expression of Mcl-1 from a 4HT-inducible construct was sufficient to prevent their death. Two independent clones of WT FDM cells were generated that each expressed either Mcl-1 or Bcl-2 following exposure to 4HT (Fig 6A). Both inducible clones expressed similar levels of Bcl-2, but the Mcl-1 clone 2 cells expressed more Mcl-1 than clone 1 cells. The over-expression of Mcl-1 had only a modest protective effect and was less effective than Bcl-2 over-expression in preventing CHX-induced cell death (Fig 6B). These results indicate that the over-expression of Mcl-1 or Bcl-2 can at least partially prevent the effect of CHX-triggered cell death.

thumbnail
Fig 6. Mcl-1 over-expression gives partial protection to wild-type cells from CHX-induced cell death.

A) Clones of WT FDM cells bearing inducible Mcl-1 or Bcl-2 expression constructs were treated for 24 h with or without 100 nM 4HT to induce Mcl-1 or Bcl-2. Cell lysates were prepared and immunoblotted with anti-Mcl-1, anti-flag (to detect flag-Bcl-2) or ß-actin as indicated. B) The same cells as in (A) were treated with 100 nM 4HT for 24 h before being culture in the presence or absence of 20 μg/ml CHX for a further 24 h before cell viabilities were determined by PI exclusion and flow cytometry. Data shown is expressed as mean ± SEM from 3 independent experiments. The numbers after a cell line’s name refer to independent clonal lines of the same genotype.

https://doi.org/10.1371/journal.pone.0164003.g006

These results indicate that cell death following addition of CHX is dependent on either Bax or Bak. To determine which of these proteins played the greater role, we generated FDM cells from mice in which the genes for either Bax or Bak were deleted. Consistent with previous results all four independent WT FDM cell lines were highly sensitive to CHX, with a significant loss in viability observed after 24 h treatment (Fig 7). Again both bax/bak DKO cell lines were resistant to CHX treatment. Interestingly, bax SKO cells were as sensitive to CHX treatment as WT cells. However, bak SKO cells showed marked resistance to CHX induced cell death, with 60% of cells remaining viable after 24 h CHX treatment. These results indicate that Bak, rather than Bax, plays the major role in FMD cell death following CHX treatment.

thumbnail
Fig 7. Bak deficient FDM cells are resistant to CHX-induced apoptosis.

Cell viabilities of WT, bax/bak DKO, and bax and bak SKO FDM cell lines were determined after 24 h of treatment with 20 μg/ml CHX. Data shown is expressed as mean ± SEM from 3 independent experiments. The numbers after a cell line’s name refer to independent clonal lines of the same genotype.

https://doi.org/10.1371/journal.pone.0164003.g007

To further test the dependence of the FDM cells on Mcl-1, we determined the effect of antagonizing Mcl-1 by inducing expression of the engineered BH3-only protein, BimS2A, which is highly selective for Mcl-1 [14]. The addition of 4HT to bax/bak DKO cells bearing a 4HT-inducible construct resulted in a robust expression of the BimS2A peptide (Fig 8A) yet, as expected, it was unable to trigger death in these cells (Fig 8B). In contrast induction of BimS2A in WT cells caused all the cells to die, indicating that the functional inactivation of Mcl-1 is sufficient to induce FDM cell death.

thumbnail
Fig 8. BimS2A induces efficient cell death in wild-type FDM cells.

A) Inducible bax/bak DKO FDM cells were treated for 24 h with or without 100 nM 4HT to induce BimS2A expression as indicated. Cell lysates were prepared and immunoblotted with anti-flag to detect flag-BimS2A or ß-actin as indicated. B) Inducible WT and bax/bak DKO FDM cells were treated with or without 100 nM 4HT to induce BimS2A expression as indicated. Cell viabilities were determined 24 h after the addition of 4HT. Data shown is expressed as mean ± SEM from 3 independent experiments.

https://doi.org/10.1371/journal.pone.0164003.g008

Discussion

It has generally been assumed that initiation of Bax/Bak dependent apoptosis requires the production or activation of one of more of the BH3-only members of the Bcl-2 family [10]. The activation of BH3-only proteins has been shown to occur via transcriptional or post-translational mechanisms in response to a number of external and intracellular signals. For example, DNA damage can promote stabilization of p53, which directly transactivates the puma locus [1517], Bid can be activated by proteolytic cleavage [18], and Bim can be regulated by phosphorylation [1922]. Once active BH3-only proteins are produced, they are believed to trigger Bax and Bak in either of two ways. The “direct activators” such as Bim, Puma and Bid can directly bind to Bax and/or Bak and cause them to form multimers in the mitochondrial outer membrane that allow exit of proteins including cytochrome c. Alternatively, the “indirect activators” such as Bad, Hrk, Bmf, and Bik bind to the anti-apoptotic Bcl-2 family members, and thereby prevent them from binding to and inhibiting Bax and Bak, and perhaps also by displacing Bim, Puma or Bid from the anti-apoptotic Bcl-2 family members so they are free to activate Bax or Bak. By either of these scenarios it is widely assumed that the BH3-only proteins perform an obligatory role in Bax/Bak activation [8, 10]. Consistent with this, mice deficient in Bim, Bid and Puma phenotypically resemble bax/bak DKO mice, suggesting they are essential for Bax/Bak activation [9].

Here we provide evidence that apoptosis can be triggered in another way, in the absence of BH3-only proteins Blk, Bad, Hrk, or Noxa, and in cells where none of the known direct activator BH3-only proteins (Bid, Bim, Puma) are present at detectable levels. Our results in IL-3 dependent myeloid lines suggest that without triggering by BH3-only proteins, Bax and Bak can spontaneously activate when levels of anti-apoptotic Bcl-2 family proteins decline. Consistent with our findings, Muer et al. reported that expression of p14ARF could induce Bak dependent apoptosis of DU145 prostate carcinoma cells by down regulation of Mcl-1 and Bcl-xl without the activation of BH3-only proteins [23]. Similarly, Chen and colleagues showed in MEFs that Bax and Bak auto-activated in the absence of BH3-only proteins when Bcl-2, Bcl-xl and Mcl-1 levels decreased following treatment with etoposide [24]. Moreover, in a tour de force involving simultaneous mutation of the genes for eight BH3-only proteins, five anti-apoptotic Bcl-2 family proteins, as well as Bax and Bak in HCT116 colorectal carcinoma cells, O'Neill et al. showed that Bax or Bak are able to associate with the outer mitochondrial membrane, activate, and cause apoptosis spontaneously, without being activated by any BH3-only protein [25].

CHX-induced death of mouse FDM cells required Bax and/or Bak, because cells lacking both Bax and Bak were profoundly resistant to CHX (although even bax/bak DKO FDM cells do eventually die if CHX is never removed from their media). Cells deficient in bak only were highly resistant to CHX-induced apoptosis, but not as resistant as bax/bak deficient cells. In contrast, bax deficient FDM cells were as sensitive to the effects of CHX as WT cells. These results indicate that CHX-triggered apoptosis of these cells is predominantly mediated by Bak, and in its absence Bax can only induce limited amounts of cell death.

These results are consistent with studies using baby mouse kidney (BMK) cells, and cells that lacked Bax and Bak [26], which were also highly resistant to CHX. Interestingly, bax deficient BMK cells that retained one allele of bak were as sensitive to CHX as WT cells, yet bak deficient cells that retained one copy of bax were as resistant to CHX as cells that lacked both bax and bak [26] indicating that Bak is the main mediator of death in these cells, as it was in the FDMs.

The common requirement for Bak in CHX-induced death in both FDM and BMK cells suggested that a similar mode of activation might occur in the two cell types. However, BMK cells that lacked Blk (Bik in humans) were also highly resistant to CHX indicating Blk is required for Bak activation in these cells [26]. A similar essential requirement for Bim in CHX-induced death has also been demonstrated in HeLa cells [27]. In contrast to these studies none of the BH3-only proteins were found to be necessary for CHX-induced FDM cell death. None of the SKO lines that lacked a single member of the BH3-only family exhibited any resistance towards CHX. Similarly, none of the DKO lines that were deficient in bad/bim, bid/bim or puma/noxa displayed any resistance towards CHX. Complementing these findings, roscovitine also lead to cell death in a bax/bak dependent manner, resulted in the loss of Mcl-1, and the BH3-only SKO and DKO cell lines did not show any protection against cell death. Using this eukaryotic model, it was not possible to knockout all the BH3-only proteins simultaneously, however the SKO and DKO lines tested do indicate that CHX is not inducing cell death through any single known BH3-only protein. Furthermore, the presence of CHX would prevent the synthesis of new BH3-only proteins (such as Puma or Noxa) to activate Bak to cause cell death. This finding supports a model in which several known BH3-only proteins are not involved in CHX-induced FDM cell death. This finding is further supported by our results using roscovitine which, like CHX, caused loss of Mcl-1 and induced Bax/Bak dependent cell death. Moreover, the finding that none of the BH3-only knockout cell lines provided protection to roscovitine treatment suggests that known BH3-only proteins are also not involved in roscovitine-induced FDM cell death.

Our results suggest that as Mcl-1 levels decline, Bax/Bak can activate spontaneously, without being activated by a ‘direct’ activator BH3-only protein, such as Bim, Puma or Bid. While it has been reported that over-expression of Bax or Bak in yeasts and mammalian cells can lead to their spontaneous activation [2830], in our experiments Bax and Bak existed at their physiological levels.

Mcl-1 is unique among members of the Bcl-2 pro-survival family in that it is rapidly turned over by the action of ubiquitin ligases (such as Mule) [3133] and can be stabilised by the action of deubiquitinases (e.g. USP9X) [34]. Consistent with this, Mcl-1 abundance was rapidly decreased to barely detectable levels in FDM cells within 4 h of CHX addition, whereas none of the other Bcl-2 family members showed any appreciable decrease over the time-course examined. Furthermore, the treatment with the proteasome inhibitor, MG132, increased abundance of Mcl-1 in resting cells and prevented the loss of Mcl-1 following CHX addition. Thus the normal proteasomal turnover of Mcl-1 can account for the loss of Mcl-1 following inhibition of protein synthesis with CHX.

Bak is predominantly inhibited by the actions of Mcl-1 and Bcl-xL and not other pro-survival family members [26, 35]. Thus Mcl-1 loss in FDM cells following protein synthesis inhibition is consistent with the observed requirement of Bak to mediate CHX-induced cell death. However, it is worth noting the bak deficient FDM cells were not completely resistant to CHX like bax/bak deficient cells. Over-expression of Mcl-1 and Bcl-2 both increased cell survival after CHX-treatment. Since Mcl-1 is rapidly turned over, its over-expression did not raise its levels as much as over-expression of Bcl-2, which is more stable, and hence over-expression of Bcl-2 gave a greater level of protection (Fig 6).

Loss of Mcl-1 has been observed previously in HeLa cells [27] and in other cell types including leukemic cells and fibroblasts [36] following inhibition of protein synthesis. However, in contrast to the results here, it was concluded in these studies that Mcl-1 loss was not the sole event to trigger cell death [27, 36]. Indeed, in HeLa cells Bim was also required for CHX-induced cell death [27] and Bim can inhibit all pro-survival family proteins [37]. Furthermore, in CHX treatment of BMK cells it was shown that Blk was required to antagonise the functions of Mcl-1 and Bcl-xL to cause death [26].

The results presented here are consistent with a model in which a decline in Mcl-1 levels is the key event leading to Bak activation and cell death following protein synthesis inhibition. This model predicts that the specific inactivation of Mcl-1 should be sufficient to cause cell death. In support of this the over-expression of BimS2A, which specifically binds and inhibits the function of Mcl-1, efficiently induced cell death in WT but not bax/bak deficient FDM cells.

In conclusion none of the tested BH3-only proteins were found to be required for CHX-induced or roscovitine-induced FDM cell death. Rather the loss of Mcl-1 was the key event leading to Bak activation and cell death. The results of this study are consistent with a model in which there is no obligatory requirement for the direct activation of Bak by several known BH3-only proteins in CHX-induced apoptosis in FDM cells.

Materials and Methods

Antibodies and chemicals

Mouse monoclonal anti-flag (M2 clone; #F1804) and anti-ß-actin (clone AC-15; #A1978) were purchased from Sigma (Castle Hill, NSW). Rabbit polyclonal anti-Mcl-1 (#600-401-394) was purchased from Rockland (Jomar Bioscience, Welland, SA). Hamster monoclonal anti-Bcl-2 (clone 3F11) was obtained from Andreas Strasser (WEHI). Anti-Bcl-w and anti-Bcl-xL were obtained from Lorraine O'Reilly (WEHI) and rat anti-A1 (clone 6D6) was provided by David Huang (WEHI). Cycloheximide (#C7698), 4-hydroxytamoxifen (4HT; #H7904), roscovitine (#R7772) and MG132 (#C2211) were purchased from Sigma.

Plasmids and cDNAs

MmMcl-1 and HsBcl-2 cDNAs were generously provided by David Huang (WEHI) while BimS2A cDNA was provided by Doug Fairlie (WEHI). BamHI/XbaI fragments or compatible fragments containing their respective coding regions were subcloned in frame into the 4HT-inducible vector, pF-5xUAS-flag-W-SV40puro [38], to generate N-terminally flag-tagged constructs.

Cell Culture

Generation of IL-3 dependent cell lines.

Factor dependent myeloid (FDM) cells were generated from E14.5 mice, by co-culturing single cell suspensions from fetal liver with fibroblasts producing a HoxB8 expressing retrovirus in the presence of 0.25 ng/ml recombinant mouse IL-3 (R&D Systems, Sapphire Biosciences, Redfern, NSW). Noxa-/-, Bmf-/-, Hrk-/-, Bik-/- and Puma-/-;Noxa-/- mice have been described before [3943]. All mice were derived from C57BL/6 embryonic stem (ES) cells or from 129SV-derived ES cells and back- crossed onto a C57BL/6 background for at least 10 generations. Cell lines were tested for IL-3 dependence by determining their ability to proliferate in the absence of IL-3 in soft agar. No cell lines were able to generate colonies under such conditions. Wild-type FDM cells and Bad-/-, Bid-/-, Bim-/-, Puma-/-, Bax-/-, Bak-/-, Bax-/-;Bak-/-, Bad-/-;Bim-/- and Bid-/-;Bim-/- FDM cells have all been described previously [2, 4]. All cells were cultured in DMEM (low glucose; Life Technologies, Melbourne, VIC) supplemented with 10% fetal bovine serum (Life Technologies), 0.25 ng/ml IL-3, penicillin G 50 U/ml/streptomycin (50 μg/ml), L-glutamine (2 mM) in a humidified atmosphere of 10% CO2 at 37°C. For IL-3 withdrawal experiments cells were washed at least 3 times with phosphate buffered saline (PBS) before being resuspended in complete media without IL-3.

Generation of 4-hydroxytamoxifen-inducible cell lines.

Lentiviruses were first prepared by transfecting 293T cells with the appropriate lentiviral vector together with pCMV-ΔR8 and pVSV-G packaging constructs using Effectene as described previously [38]. 48 h after transfection viral supernatants were filtered, mixed with polybrene (4 μg/mL), and added to target cells which were then centrifuged at 1 250 g for 90 min at room temperature. Stably infected 4HT-inducible cell lines were selected in the presence of puromycin and hygromycin B. Gene expression in target cells was induced by the addition of 100 nM 4HT.

Cell viability assays

Cells were centrifuged at 1 700 rpm (415 g) for 5 min at 4°C and re-suspended in 100–200 μl PBS containing 1 μg/ml of propidium iodide (PI) and sorted using a FACSCalibur flow cytometer (Becton Dickinson, North Ryde, New South Wales). Twenty thousand events per sample were collected and analyzed using WEASEL software (version 2.2.2, WEHI). Viable cells were identified by their ability to exclude propidium iodide. Unless otherwise stated, all cell death/viability experiments were independently repeated at least three times.

Cell lysis and immunoblotting

Cells were centrifuged at 1 700 rpm for 5 min and washed with ice-cold PBS. Cells were lysed directly in sample buffer (100 mM Tris-Cl pH 6.8, 4% (w/v) SDS, 20% (v/v) glycerol) and boiled for 10 min. Alternatively, cells were re-suspended in DISC lysis buffer [44] supplemented with complete protease inhibitor cocktail (Roche), 10 mM NaF, 2 mM Na pyrophosphate, 1 mM Na molybdate and 5 mM ß-glycerophosphate, incubated on ice for 30–60 min and clarified by centrifugation at 13 000 g for 10 min at 4°C. All lysates were stored at -80°C. Clarified lysates (50 μg) were mixed with sample buffer, boiled and separated by sodium dodecyl sulphate polyacrylamide gel electrophoresis (SDS-PAGE) on linear gels or 4–20% Tris-glycine gradient gels (Bio-Rad, Gladesville, NSW) or 4–12% Bis-Tris gradient NuPage gels (Life Technologies) and transferred to Hybond-C nitrocellulose membrane (GE, Castle Hill, NSW). Membranes were blocked in 5% skim milk powder in Tris-buffered saline containing 0.1% Tween-20 (TBST) or PBS containing 0.1% Tween-20 (PBST) and incubated with primary antibody overnight at 4°C or for 2 hours at room temperature. Membranes were washed four times with TBST or PBST at room temperature, incubated with horseradish peroxidase conjugated secondary antibody (GE) and washed before detection with enhanced chemiluminescence. Where necessary, membranes were stripped in buffer (100 mM Tris adjusted to pH 7.4, 2% SDS, 350 μl/50 ml β-mercaptoethanol) incubated for 15 min at 60°C. The membrane was rinsed thoroughly with dH2O (3 x 20 ml) then rinsed three times with TBST before being re-blocked with milk.

Acknowledgments

This work was supported by an Association of International Cancer Research project grant (09–0084) and Cancer Council of WA project grants (1010749 and 1067515) and fellowship to BAC, and National Health and Medical Research Council (Canberra, Australia) Project grant (384404), fellowship (1016701), and Program Grant (461221) to DLV, and operational infrastructure grants through the Australian Government IRISS and the Victorian State Government OIS. We generously thank Andreas Strasser for providing Bcl-2 antibody, Lorraine O'Reilly for providing Bcl-w and Bcl-xL antibodies, David Huang for providing A1 antibody and Mcl-1 and Bcl-2 cDNAs and Doug Fairlie for providing BimS2A cDNA.

Author Contributions

  1. Conceptualization: KJG IEG PGE DLV BAC JEV.
  2. Formal analysis: KJG BAC DLV.
  3. Funding acquisition: BAC DLV.
  4. Investigation: KJG MLFE JvV GCY BAC DLV.
  5. Methodology: KJG IEG PGE DLV BAC JEV.
  6. Project administration: BAC DLV.
  7. Resources: PGE DLV BAC.
  8. Supervision: BAC DLV.
  9. Visualization: KJG BAC DLV.
  10. Writing – original draft: BAC KJG PGE DLV.
  11. Writing – review & editing: KJG PGE DLV BAC.

References

  1. 1. Vaux DL, Cory S, Adams JM. Bcl-2 gene promotes haemopoietic cell survival and cooperates with c-myc to immortalize pre-B cells. Nature. 1988;335(6189):440–2. pmid:3262202
  2. 2. Ekert PG, Jabbour AM, Manoharan A, Heraud JE, Yu J, Pakusch M, et al. Cell death provoked by loss of Interleukin-3 signalling is independent of Bad, Bim, and PI3 Kinase, but depends in part on Puma. Blood. 2006;108:1461–8. pmid:16705087
  3. 3. Ekert PG, Read SH, Silke J, Marsden VS, Kaufmann H, Hawkins CJ, et al. Apaf-1 and caspase-9 accelerate apoptosis, but do not determine whether factor-deprived or drug-treated cells die. J Cell Biol. 2004;165(6):835–42. pmid:15210730
  4. 4. Jabbour AM, Heraud JE, Daunt CP, Kaufmann T, Sandow J, O'Reilly LA, et al. Puma indirectly activates Bax to cause apoptosis in the absence of Bid or Bim. Cell Death Differ. 2009;16(4):555–63. pmid:19079139
  5. 5. Perkins AC, Cory S. Conditional immortalization of mouse myelomonocytic, megakaryocytic and mast cell progenitors by the Hox-2.4 homeobox gene. Embo J. 1993;12(10):3835–46. pmid:8104786
  6. 6. Gilden RV, Carp RI. Effects of cycloheximide and puromycin on synthesis of simian virus 40 T antigen in green monkey kidney cells. J Bacteriol. 1966;91(3):1295–7. pmid:4286450
  7. 7. Vaux DL, Weissman IL. Neither macromolecular synthesis nor myc is required for cell death via the mechanism that can be controlled by bcl 2. Molecular and Cellular Biology. 1993;13(11):7000–5. pmid:7692234
  8. 8. Llambi F, Moldoveanu T, Tait SW, Bouchier-Hayes L, Temirov J, McCormick LL, et al. A unified model of mammalian BCL-2 protein family interactions at the mitochondria. Mol Cell. 2011;44(4):517–31. pmid:22036586
  9. 9. Ren D, Tu HC, Kim H, Wang GX, Bean GR, Takeuchi O, et al. BID, BIM, and PUMA are essential for activation of the BAX- and BAK-dependent cell death program. Science. 2010;330(6009):1390–3. pmid:21127253
  10. 10. Czabotar PE, Lessene G, Strasser A, Adams JM. Control of apoptosis by the BCL-2 protein family: implications for physiology and therapy. Nat Rev Mol Cell Biol. 2014;15(1):49–63. pmid:24355989
  11. 11. Stewart DP, Koss B, Bathina M, Perciavalle RM, Bisanz K, Opferman JT. Ubiquitin-independent degradation of antiapoptotic MCL-1. Molecular and cellular biology. 2010;30(12):3099–110. pmid:20385764
  12. 12. De Biasio A, Vrana JA, Zhou P, Qian L, Bieszczad CK, Braley KE, et al. N-terminal truncation of antiapoptotic MCL1, but not G2/M-induced phosphorylation, is associated with stabilization and abundant expression in tumor cells. The Journal of biological chemistry. 2007;282(33):23919–36. pmid:17561513
  13. 13. MacCallum DE, Melville J, Frame S, Watt K, Anderson S, Gianella-Borradori A, et al. Seliciclib (CYC202, R-Roscovitine) induces cell death in multiple myeloma cells by inhibition of RNA polymerase II-dependent transcription and down-regulation of Mcl-1. Cancer research. 2005;65(12):5399–407. pmid:15958589
  14. 14. Lee EF, Czabotar PE, van Delft MF, Michalak EM, Boyle MJ, Willis SN, et al. A novel BH3 ligand that selectively targets Mcl-1 reveals that apoptosis can proceed without Mcl-1 degradation. The Journal of cell biology. 2008;180(2):341–55. pmid:18209102
  15. 15. Han JW, Flemington C, Houghton AB, Gu ZM, Zambetti GP, Lutz RJ, et al. Expression of bbc3, a pro-apoptotic BH3-only gene, is regulated by diverse cell death and survival signals. Proc Natl Acad Sci U S A. 2001;98(20):11318–23. pmid:11572983
  16. 16. Nakano K, Vousden KH. PUMA, a novel proapoptotic gene, is induced by p53. Mol Cell. 2001;7(3):683–94. pmid:11463392
  17. 17. Yu J, Zhang L, Hwang PM, Kinzler KW, Vogelstein B. PUMA induces the rapid apoptosis of colorectal cancer cells. Mol Cell. 2001;7(3):673–82. pmid:11463391
  18. 18. Li HL, Zhu H, Xu CJ, Yuan JY. Cleavage of bid by caspase 8 mediates the mitochondrial damage in the fas pathway of apoptosis. Cell. 1998;94(4):491–501. pmid:9727492
  19. 19. Lei K, Davis RJ. JNK phosphorylation of Bim-related members of the Bcl2 family induces Bax-dependent apoptosis. Proc Natl Acad Sci U S A. 2003;100:2432–7. pmid:12591950
  20. 20. Ley R, Balmanno K, Hadfield K, Weston CR, Cook SJ. Activation of the ERK1/2 signalling pathway promotes phosphorylation and proteasome-dependent degradation of the BH3-only protein Bim. J Biol Chem. 2003;278:18811–6. pmid:12646560
  21. 21. Luciano F, Jacquel A, Colosetti P, Herrant M, Cagnol S, Pages G, et al. Phosphorylation of Bim-EL by Erk1/2 on serine 69 promotes its degradation via the proteasome pathway and regulates its proapoptotic function. Oncogene. 2003;22(43):6785–93. pmid:14555991
  22. 22. Putcha GV, Le S, Frank S, Besirli CG, Clark K, Chu B, et al. JNK-Mediated BIM Phosphorylation Potentiates BAX-Dependent Apoptosis. Neuron. 2003;38(6):899–914. pmid:12818176
  23. 23. Muer A, Overkamp T, Gillissen B, Richter A, Pretzsch T, Milojkovic A, et al. p14(ARF)-induced apoptosis in p53 protein-deficient cells is mediated by BH3-only protein-independent derepression of Bak protein through down-regulation of Mcl-1 and Bcl-xL proteins. J Biol Chem. 2012;287(21):17343–52. pmid:22354970
  24. 24. Chen HC, Kanai M, Inoue-Yamauchi A, Tu HC, Huang Y, Ren D, et al. An interconnected hierarchical model of cell death regulation by the BCL-2 family. Nature cell biology. 2015;17(10):1270–81. pmid:26344567
  25. 25. O'Neill KL, Huang K, Zhang J, Chen Y, Luo X. Inactivation of prosurvival Bcl-2 proteins activates Bax/Bak through the outer mitochondrial membrane. Genes Dev. 2016;30(8):973–88. pmid:27056669
  26. 26. Shimazu T, Degenhardt K, Nur EKA, Zhang J, Yoshida T, Zhang Y, et al. NBK/BIK antagonizes MCL-1 and BCL-XL and activates BAK-mediated apoptosis in response to protein synthesis inhibition. Genes Dev. 2007;21(8):929–41. pmid:17403773
  27. 27. Adams KW, Cooper GM. Rapid turnover of mcl-1 couples translation to cell survival and apoptosis. The Journal of biological chemistry. 2007;282(9):6192–200. pmid:17200126
  28. 28. Chittenden T, Harrington EA, Oconnor R, Flemington C, Lutz RJ, Evan GI, et al. Induction of apoptosis by the bcl-2 homologue bak. Nature. 1995;374(6524):733–6. pmid:7715730
  29. 29. Dewson G, Kratina T, Sim HW, Puthalakath H, Adams JM, Colman PM, et al. To trigger apoptosis, Bak exposes its BH3 domain and homodimerizes via BH3:groove interactions. Mol Cell. 2008;30(3):369–80. pmid:18471982
  30. 30. Ink B, Zornig M, Baum B, Hajibagheri N, James C, Chittenden T, et al. Human bak induces cell death in schizosaccharomyces pombe with morphological changes similar to those with apoptosis in mammalian cells. Molecular & Cellular Biology. 1997;17(5):2468–74.
  31. 31. Nijhawan D, Fang M, Traer E, Zhong Q, Gao W, Du F, et al. Elimination of Mcl-1 is required for the initiation of apoptosis following ultraviolet irradiation. Genes Dev. 2003;17(12):1475–86. pmid:12783855
  32. 32. Zhao Y, Altman BJ, Coloff JL, Herman CE, Jacobs SR, Wieman HL, et al. Glycogen synthase kinase 3alpha and 3beta mediate a glucose-sensitive antiapoptotic signaling pathway to stabilize Mcl-1. Molecular and Cellular Biology. 2007;27(12):4328–39. pmid:17371841
  33. 33. Zhong Q, Gao W, Du F, Wang X. Mule/ARF-BP1, a BH3-only E3 ubiquitin ligase, catalyzes the polyubiquitination of Mcl-1 and regulates apoptosis. Cell. 2005;121(7):1085–95. pmid:15989957
  34. 34. Schwickart M, Huang X, Lill JR, Liu J, Ferrando R, French DM, et al. Deubiquitinase USP9X stabilizes MCL1 and promotes tumour cell survival. Nature. 2010;463(7277):103–7. pmid:20023629
  35. 35. Willis SN, Chen L, Dewson G, Wei A, Naik E, Fletcher JI, et al. Proapoptotic Bak is sequestered by Mcl-1 and Bcl-xL, but not Bcl-2, until displaced by BH3-only proteins. Genes Dev. 2005;18:18.
  36. 36. Lindqvist LM, Vikstrom I, Chambers JM, McArthur K, Ann Anderson M, Henley KJ, et al. Translation inhibitors induce cell death by multiple mechanisms and Mcl-1 reduction is only a minor contributor. Cell Death Dis. 2012;3:e409. pmid:23059828
  37. 37. Chen L, Willis SN, Wei A, Smith BJ, Fletcher JI, Hinds MG, et al. Differential Targeting of Prosurvival Bcl-2 Proteins by Their BH3-Only Ligands Allows Complementary Apoptotic Function. Mol Cell. 2005;17(3):393–403. pmid:15694340
  38. 38. Callus BA, Moujallad DM, Silke J, Gerl R, Jabbour AM, Ekert PG, et al. Triggering of apoptosis by Puma is determined by the threshold set by prosurvival Bcl-2 family proteins. J Mol Biol. 2008;384(2):313–23. pmid:18835564
  39. 39. Coultas L, Bouillet P, Stanley EG, Brodnicki TC, Adams JM, Strasser A. Proapoptotic BH3-only Bcl-2 family member Bik/Blk/Nbk is expressed in hemopoietic and endothelial cells but is redundant for their programmed death. Molecular and Cellular Biology. 2004;24(4):1570–81. pmid:14749373
  40. 40. Coultas L, Terzano S, Thomas T, Voss A, Reid K, Stanley EG, et al. Hrk/DP5 contributes to the apoptosis of select neuronal populations but is dispensable for haematopoietic cell apoptosis. J Cell Sci. 2007;120(Pt 12):2044–52. pmid:17535852
  41. 41. Labi V, Erlacher M, Kiessling S, Manzl C, Frenzel A, O'Reilly L, et al. Loss of the BH3-only protein Bmf impairs B cell homeostasis and accelerates gamma irradiation-induced thymic lymphoma development. J Exp Med. 2008;205(3):641–55. pmid:18299399
  42. 42. Michalak EM, Villunger A, Adams JM, Strasser A. In several cell types tumour suppressor p53 induces apoptosis largely via Puma but Noxa can contribute. Cell Death Differ. 2008;15(6):1019–29. pmid:18259198
  43. 43. Villunger A, Michalak EM, Coultas L, Mullauer F, Bock G, Ausserlechner MJ, et al. p53- and drug-induced apoptotic responses mediated by BH3-only proteins puma and noxa. Science. 2003;302(5647):1036–8. pmid:14500851
  44. 44. Muzio M, Chinnaiyan AM, Kischkel FC, Orourke K, Shevchenko A, Ni J, et al. Flice, a novel fadd-homologous ice/ced-3-like protease, is recruited to the cd95 (fas/apo-1) death-inducing signaling complex. Cell. 1996;85(6):817–27. pmid:8681377