The pro-apoptotic BCL-2 protein BAX commits human cells to apoptosis by permeabilizing the outer mitochondrial membrane. BAX activation has been suggested to require the separation of helix α5 from α6 – the ‘latch’ from the ‘core’ domain – among other conformational changes. Here, we show that conformational changes in this region impair BAX translocation to the mitochondria and retrotranslocation back into the cytosol, and therefore BAX inhibition, but not activation. Redirecting misregulated BAX to the mitochondria revealed an alternative mechanism of BAX inhibition. The E3 ligase parkin, which is known to trigger mitochondria-specific autophagy, ubiquitylates BAX K128 and targets the pro-apoptotic BCL-2 protein for proteasomal degradation. Retrotranslocation-deficient BAX is completely degraded in a parkin-dependent manner. Although only a minor pool of endogenous BAX escapes retrotranslocation into the cytosol, parkin-dependent targeting of misregulated BAX on the mitochondria provides substantial protection against BAX apoptotic activity.

Mitochondrial apoptosis is a key form of programmed cell death (Hotchkiss et al., 2009). The activities of the BCL-2 proteins BAX and BAK commit mammalian cells to apoptosis by inducing outer mitochondrial membrane (OMM) permeabilization. The release of cytochrome c (cyt c) and other proteins from the mitochondrial intermembrane space into the cytosol initiates the caspase cascade, efficiently dismantling the cell (Martinou and Youle, 2011; Tait and Green, 2010). Although cells can survive limited mitochondrial permeabilization (Ichim et al., 2015), activation of BAX and BAK is usually the first irreversible step in mitochondrial apoptosis signaling (Youle and Strasser, 2008). Consequently, BAX and BAK are regulated by a complex network of protein interactions both within and outside the BCL-2 family. BAX and BAK contain three BCL-2 homology domains (BH1–BH3) and their primary antagonists are pro-survival BCL-2 proteins such as BCL-2 itself, BCL-xL (BCL2L1) and MCL-1, which have four BH domains (BH1–BH4). All these proteins are structurally similar and contain a C-terminal transmembrane domain (TMD) mediating association with mitochondria to different extents.

BAX and BAK specifically target the voltage-dependent anion channel 2 (VDAC2) on the OMM, forming large protein complexes (Cheng et al., 2003; Lauterwasser et al., 2016; Lazarou et al., 2010; Ma et al., 2014). Pro-survival BCL-2 proteins antagonize OMM localization and activation of BAX by constant retrotranslocation of mitochondrial BAX into the cytosol (Edlich et al., 2011). Permanent translocation and retrotranslocation establish an equilibrium between cytosolic and mitochondrial BAX that regulates BAX and thus BAX-dependent OMM permeabilization (Edlich, 2015; Edlich et al., 2011; Schellenberg et al., 2013; Todt et al., 2013, 2015). BH3-only proteins (containing only one BH motif) can trigger mitochondrial BAX accumulation by inhibition of pro-survival BCL-2 proteins and are also thought to induce active BAX conformations by direct interaction (Edlich et al., 2011; Letai et al., 2002; Llambi et al., 2011; Willis et al., 2005, 2007). The functionally redundant BAK is controlled by the same process, but different shuttling rates dependent on the C-terminal TMDs result in primarily cytosolic BAX and predominantly mitochondrial BAK (Edlich, 2015; Todt et al., 2015). Exposure of the TMD plays an important role in BAX activation (Annis et al., 2005; George et al., 2007; Nechushtan et al., 1999; Suzuki et al., 2003). However, the structural analysis of BAX lacking the TMD (helix α9) after induction of conformational changes by various means also suggests the separation of the ‘latch’ (helix α6, α7 and α8) from the ‘core’ domain (helices α1–α5) during BAX activation (Czabotar et al., 2013).

Inhibition of BAX has also been suggested to occur independently of BCL-2 proteins. Cytosolic interactions between BAX and Ku70 (also known as XRCC6), 14-3-3 isoforms (Samuel et al., 2001; Tsuruta et al., 2004), Pin-1 (Shen et al., 2009) and other proteins have been implicated in BAX regulation. Recently, the E3 ligase parkin has been suggested to ubiquitylate the BAX BH3 domain, thereby targeting cytosolic BAX to proteasomal degradation prior to translocation to the mitochondria (Charan et al., 2014; Johnson et al., 2012). However, the comprehensive analysis of cytosolic BAX and its potential interactions revealed that cytosolic BAX interactions are not apparent (Hsu and Youle, 1998; Vogel et al., 2012). Nevertheless, the importance of BAX activity in particular for the commitment to apoptosis induced by a variety of apoptotic stresses (Wang and Youle, 2011) suggests additional layers of BAX-specific regulation.

Tethering helix α5 to α6 blocks the association of BAX with mitochondria

The structure of active BAX has been subject to extensive research. An intramolecular disulfide tether between helix α5 and α6 (V121C/I136C) was shown to interfere with BAX activity in organellar cyt c release assays, suggesting that separation of helix α5 from helix α6 occurs during BAX activation (Czabotar et al., 2013). We studied the importance of conformational changes separating helix α5 and α6 by introducing this disulfide tether between BAX V121C and I136C (BAX 5-6) expressed in HCT116 BAX–BAK double knockout (DKO) cells (Fig. 1A). This approach parallels our previous analysis of BAX 1-2/L-6 in the context of living cells, leading to the discovery of BAX retrotranslocation from the mitochondria into the cytosol (Edlich et al., 2011). Similar to BAX 1-2/L-6, the engineered disulfide bond between helix α5 and α6 is largely oxidized and shielded from the reducing environment of the cytosol, as shown by altered SDS-PAGE migration and the lack of Mal–PEG labeling for the vast majority of BAX 5-6 also in denaturing conditions (Fig. 1B and Fig. S1A).

Fig. 1.

Tethering helix α5 to α6 blocks association of BAX to mitochondria. (A) Three-dimensional structure of BAX [Protein data bank (PDB) accession no., 1F16] containing V121C and I136C substitutions (5-6, red sticks) to form disulfide bonds constraining α-helices 5 (H5, orange) and 6 (H6, green) created using Pymol software (DeLano Scientific LLC). BH3 domain (BH3) and transmembrane domain (TMD) are depicted in blue and cyan, respectively. (B) Reduced SH groups of BAX 5-6, wild-type BAX and BAX C62/126S (ΔSH, without cysteines) expressed in HCT116 BAX–BAK DKO cells were detected by Mal–PEG labeling (+10 kDa per SH group) and analyzed by SDS-PAGE and western blot using anti-Myc antibody. BAX 5-6 remains largely unlabeled, suggesting formation of the α5–α6 tether. (C) Western blot analysis of BAX ΔSH and BAX 5-6 localization. Cytosol (C) and heavy membrane fraction (HM) of DKO cells are displayed. AKT1 and Tom20 serve as fractionation controls. See also Fig. S2. (D) Confocal images of DKO cells expressing wild-type BAX or BAX 5-6. GFP fluorescence of the expressed protein variants is depicted on the left and in the merge in the right panels in green. Mitochondria are stained with MitoTracker Red. (E) BN-PAGE analysis of isolated mitochondria from DKO cells expressing wild-type BAX (WT), BAX 5-6, BAX W139A, BAX L141A and BAX ΔH6 migrated on the same gel using anti-VDAC or anti-BAX antibodies. Note that lanes shown in left and middle blots shown are duplicated from the larger blots in Fig. 3B. (F) Superimposition of the three-dimensional structures of BAX (PDB, 1F16; green) and BAK (PDB, 2IMS; cyan) depicting α-helices 6 of BAX (yellow) and BAK (blue), the BAX residues W139 and L141, and the corresponding BAK residues F157 and V159 using Pymol. (G) Subcellular localization of wild-type BAX, BAX 5-6 and BAX W139A. Cytosol (C) and heavy membrane fraction (HM) are shown. AKT1 and VDAC serve as fractionation controls. (H) FLIP measurements of BAX 5-6 or wild-type BAX [WT, similar to BAX ΔSH (Edlich et al., 2011)] show reduced GFP-BAX fluorescence in the cytosol of the targeted cells (outlined). Mitochondrial fluorescence levels (arrows; BAX WT) are too low for analysis of BAX 5-6 retrotranslocation. During FLIP measurements, mitochondrial GFP fluorescence is monitored, while the cytosol is bleached repeatedly. The neighboring cell serves as control for bleaching by imaging. Time points in seconds are displayed on top. Scale bars: 5 µm. All experiments are representative of n=3 replicates, except for H, where n=10.

Fig. 1.

Tethering helix α5 to α6 blocks association of BAX to mitochondria. (A) Three-dimensional structure of BAX [Protein data bank (PDB) accession no., 1F16] containing V121C and I136C substitutions (5-6, red sticks) to form disulfide bonds constraining α-helices 5 (H5, orange) and 6 (H6, green) created using Pymol software (DeLano Scientific LLC). BH3 domain (BH3) and transmembrane domain (TMD) are depicted in blue and cyan, respectively. (B) Reduced SH groups of BAX 5-6, wild-type BAX and BAX C62/126S (ΔSH, without cysteines) expressed in HCT116 BAX–BAK DKO cells were detected by Mal–PEG labeling (+10 kDa per SH group) and analyzed by SDS-PAGE and western blot using anti-Myc antibody. BAX 5-6 remains largely unlabeled, suggesting formation of the α5–α6 tether. (C) Western blot analysis of BAX ΔSH and BAX 5-6 localization. Cytosol (C) and heavy membrane fraction (HM) of DKO cells are displayed. AKT1 and Tom20 serve as fractionation controls. See also Fig. S2. (D) Confocal images of DKO cells expressing wild-type BAX or BAX 5-6. GFP fluorescence of the expressed protein variants is depicted on the left and in the merge in the right panels in green. Mitochondria are stained with MitoTracker Red. (E) BN-PAGE analysis of isolated mitochondria from DKO cells expressing wild-type BAX (WT), BAX 5-6, BAX W139A, BAX L141A and BAX ΔH6 migrated on the same gel using anti-VDAC or anti-BAX antibodies. Note that lanes shown in left and middle blots shown are duplicated from the larger blots in Fig. 3B. (F) Superimposition of the three-dimensional structures of BAX (PDB, 1F16; green) and BAK (PDB, 2IMS; cyan) depicting α-helices 6 of BAX (yellow) and BAK (blue), the BAX residues W139 and L141, and the corresponding BAK residues F157 and V159 using Pymol. (G) Subcellular localization of wild-type BAX, BAX 5-6 and BAX W139A. Cytosol (C) and heavy membrane fraction (HM) are shown. AKT1 and VDAC serve as fractionation controls. (H) FLIP measurements of BAX 5-6 or wild-type BAX [WT, similar to BAX ΔSH (Edlich et al., 2011)] show reduced GFP-BAX fluorescence in the cytosol of the targeted cells (outlined). Mitochondrial fluorescence levels (arrows; BAX WT) are too low for analysis of BAX 5-6 retrotranslocation. During FLIP measurements, mitochondrial GFP fluorescence is monitored, while the cytosol is bleached repeatedly. The neighboring cell serves as control for bleaching by imaging. Time points in seconds are displayed on top. Scale bars: 5 µm. All experiments are representative of n=3 replicates, except for H, where n=10.

Fractionation of cells expressing BAX 5-6 showed a shift of tethered BAX from the mitochondria to the cytosol compared with that in cells expressing BAX ΔSH (which localizes like wild-type BAX; Edlich et al., 2011) at similar expression levels (Fig. 1C,D, Fig. S1B and Fig. S2A). The voltage-dependent anion channel 2 (VDAC2) mediates mitochondrial BAX association and is essential for BAX retrotranslocation (Lauterwasser et al., 2016; Ma et al., 2014). Therefore, we tested the requirement of conformational changes separating helix α5 and α6 for VDAC2 interactions with BAX. BN-PAGE analysis shows a substantial reduction of BAX-containing VDAC2 complexes when the tether between BAX helices α5 and α6 was formed (Fig. 1E and Fig. S2B). A BAX variant lacking helix α6 also did not form complexes with VDAC2. We, therefore, hypothesized that the stabilized BAX 5-6 structure prevents interactions between BAX helix α6 and VDAC2. Therefore, alanine substitutions of the bulky hydrophobic residues W139 and L141 on both sides of helix α6, corresponding to residues F157 and V159 in the similarly regulated BAK (Fig. 1F), were tested and confirm the importance of both sides of helix α6 by impairing the formation of VDAC2-containing BAX complexes (Fig. 1E). W139A also abolishes contact between BAX and the OMM (Fig. 1G). Fluorescence loss in photobleaching (FLIP) experiments to analyze BAX 5-6 retrotranslocation from the mitochondria into the cytosol also revealed diminished mitochondrial BAX 5-6 association, rendering the measurement of BAX 5-6 retrotranslocation in comparison to the wild type impossible (Fig. 1H). The loss of cytosolic BAX fluorescence during FLIP emphasizes the significant reduction of a mitochondrial BAX pool by the α5–α6 tether (Fig. 1C,D,H). Taken together, the separation of helix α5 from α6 and thus exposure of helix α6 is important for efficient BAX association with the mitochondria in healthy cells.

BAX 5-6 can commit cells to apoptosis

Impaired mitochondrial association of BAX 5-6 could cause reduced BAX activity, as observed with mitochondrial BAX 5-6 S184L isolated from MEF cells (Czabotar et al., 2013). Surprisingly, BAX 5-6 commits HCT116 BAX/BAK DKO cells to apoptosis at least as potent as wild-type BAX (Fig. 2A). Profound BAX 5-6 activity despite reduced OMM-association of this variant could result from impaired inhibition in the absence of BAX retrotranslocation complexes. However, we cannot rule out that a minor BAX 5-6 pool lacking the α5–α6 tether (Fig. 1B) due to reduction of the involved disulfides has triggered the measured activity. Increased BAX 5-6 activity compared with the wild type is also observed by the analysis of caspase-dependent cleavage of poly (ADP-ribose) polymerase (PARP), the presence of active BAX detected by the monoclonal antibody 6A7 and the release of cyt c following apoptosis stimulation (Fig. 2B-D). Conformational changes such as the separation of BAX helices α5 and α6 are dispensable for BAX activation, which is in line with the possibility of helices α5 and α6 inserted in concert into the OMM in active BAX (Annis et al., 2005). Therefore, separation of the ‘core’ domain and ‘latch’ is important for association of BAX with mitochondria and inhibition in non-apoptotic cells, but is apparently not required for BAX activation.

Fig. 2.

BAX 5-6 commits cells to apoptosis. (A) Caspase-3/7 activity measurements of HCT116 BAX–BAK DKO cells expressing wild-type BAX or BAX 5-6 with or without 1 µM staurosporine (STS). Caspase activity is displayed in relative fluorescence units (RFU) (n≥3). (B) Western blot analysis of PARP cleavage following MOM permeabilization in the absence or the presence of 1 µM STS. Similar sample loading is controlled by actin. (C) Presence of active BAX conformation in the absence or the presence of 1 µM actinomycin D (ActD) analyzed by monoclonal anti-BAX antibody 6A7 depicted as a percentage of total BAX-expressing cells. (D) Cytochrome c (cyt c) release with or without 1 µM ActD depicted as a percentage of total BAX-expressing cells. Data represent mean±s.e.m. (n=3, unless otherwise indicated); **P≤0.01, ***P≤0.001 one-way ANOVA using the Holm–Sidak method for statistically significant differences.

Fig. 2.

BAX 5-6 commits cells to apoptosis. (A) Caspase-3/7 activity measurements of HCT116 BAX–BAK DKO cells expressing wild-type BAX or BAX 5-6 with or without 1 µM staurosporine (STS). Caspase activity is displayed in relative fluorescence units (RFU) (n≥3). (B) Western blot analysis of PARP cleavage following MOM permeabilization in the absence or the presence of 1 µM STS. Similar sample loading is controlled by actin. (C) Presence of active BAX conformation in the absence or the presence of 1 µM actinomycin D (ActD) analyzed by monoclonal anti-BAX antibody 6A7 depicted as a percentage of total BAX-expressing cells. (D) Cytochrome c (cyt c) release with or without 1 µM ActD depicted as a percentage of total BAX-expressing cells. Data represent mean±s.e.m. (n=3, unless otherwise indicated); **P≤0.01, ***P≤0.001 one-way ANOVA using the Holm–Sidak method for statistically significant differences.

Proteasomal degradation controls BAX when retrotranslocation is impaired

The demonstrated activity of BAX 5-6 suggests that HCT116 cells have the means to inhibit this BAX variant in order to survive. We reasoned that the requirement of BAX inhibition would be elevated if BAX is targeted to the OMM. BAX binding to the OMM is mediated by helix α6 and TMD interactions with VDAC2 (Lauterwasser et al., 2016). Since the hydrophobicity of the TMD determines the BAX shuttling rate (Todt et al., 2015), we introduced an S184V substitution to increase the mitochondrial pool of BAX 5-6 (Nechushtan et al., 1999; Fig. 3A). Blue native (BN)-PAGE analysis showed a vastly increased level of VDAC2-containing BAX complexes mediated by the S184V substitution (Fig. 3B). These results suggest that BAX TMD functions are not impaired by the BAX α5–α6 tether. Cells expressing BAX 5-6 S184V thus contain a pool of VDAC2 complexes as observed with wild-type BAX. These complexes of mitochondrial BAX 5-6 (and BAX 5-6 S184V) were similar in size and likely composition to the wild type and provide no rationale for increased apoptotic activity of BAX 5-6. In parallel to BAX 5-6, the apoptotic activity of BAX 5-6 S184V was significantly increased compared with the wild type in response to apoptotic stimuli (Fig. S3). Pro-apoptotic activity of BAX 5-6 S184V can also be detected in the absence of apoptotic stimuli, mirroring the properties of BAX S184V (Nechushtan et al., 1999). Confocal analysis revealed cytosolic and mitochondrial localization of BAX 5-6 S184V (Fig. 3C). Surprisingly, ectopic expression of BAX 5-6 S184V resulted in significantly lower protein levels than expected. However, in the presence of the proteasome inhibitor MG132 or the ubiquitin-activating enzyme E1 inhibitor PYR41, the levels of BAX 5-6 S184V were substantially increased (Fig. 3D,E), suggesting proteasomal degradation of misregulated BAX. By contrast, wild-type BAX and BAX 5-6 levels were affected to a much lesser extent in the presence of MG132 or PYR41 (Fig. 3F,G and Fig. S4). Therefore, proteasomal degradation represents an additional layer of BAX regulation that is dependent on mitochondrial BAX localization. Analysis of BAX 5-6 S184V retrotranslocation by FLIP measurements revealed decreased shuttling rates compared with BAX S184V (Fig. 3H,I). The S184V substitution results in a marked reduction of BAX retrotranslocation (Edlich et al., 2011; Todt et al., 2015) and this analysis was performed with ectopically expressed BCL-xL to prevent BAX activation from obscuring the results. The seemingly moderate reduction of BAX retrotranslocation by the α5–α6 tether could have a strong impact on the regulation of mitochondrial apoptosis, as BAX activation depends on the dwell time of BAX molecules on the OMM (Todt et al., 2015). BAX 5-6 S184V showed prominent ubiquitylation (Fig. 3J) whereas ubiquitylated wild-type BAX was only detectable in the presence of a ubiquitin ΔK variant. Ubiquitin ΔK is randomly incorporated into poly-ubiquitin chains in the presence of endogenous ubiquitin and stabilizes ubiquitylated protein species by less-efficient proteasomal degradation of shorter ubiquitin chains. Therefore, ubiquitylation occurs only with a mitochondrial minor pool of wild-type BAX, but this pool increases when BAX retrotranslocation is impaired. Endogenous BAX levels increased in a time-dependent manner following inhibition of the proteasome or ubiquitylation (Fig. S4C). Proteasomal BAX degradation could represent a quality control mechanism for BAX retrotranslocation, removing slowly shuttling BAX molecules from the OMM and thus preventing OMM permeabilization (Fig. 3K).

Fig. 3.

Proteasomal degradation prevents activation of misregulated BAX. (A) BAX (blue) translocation to the mitochondria targets VDAC2 (purple) in the outer mitochondrial membrane (OMM) and VDAC2 serves as platform for BAX retrotranslocation (Lauterwasser et al., 2016). BAX is retrotranslocated upon transient interactions with BCL-xL (red). Mitochondrial association of BAX is impaired by the α5 to α6 tether (bottom left). Addition of the S184V substitution increases BAX association with VDAC2 (bottom right). (B) BN-PAGE analysis of isolated mitochondria from HCT116 BAX–BAK DKO cells expressing wild-type BAX (WT), BAX 5-6, BAX 5-6 S184V or BAX S184V using anti-VDAC or anti-BAX antibodies. Note that selected WT and 5-6 sample lanes from these blots are duplicated in Fig. 1E. (C) Confocal images of DKO cells expressing BAX S184V or BAX 5-6 S184V. Mitochondrial BAX appears yellow in the merge images. (D–G) Protein levels of BAX 5-6 S184V (D,E) and WT BAX (F,G) in the absence and the presence of the proteasome inhibitor MG132 or the ubiquitin-activating enzyme E1 enzyme inhibitor PYR41 analyzed by western blotting. VDAC serves as loading control. (H) FLIP of BAX S184V (top) or BAX 5-6 S184V (bottom) in the presence of overexpressed BCL-xL diminishes GFP fluorescence in the cytoplasm of targeted cells (outlined) completely after 80 s, and GFP fluorescence is detected only on the mitochondria (arrows). (I) Retrotranslocation of BAX S184V and BAX 5-6 S184V (blue) with BCL-xL overexpression (n≥3). (J) Immunoprecipitation of HA-tagged ubiquitin (WT, wild-type; ΔK, all lysine residues substituted to arginine) from whole-cell lysates of DKO cells expressing wild-type BAX or BAX 5-6 S184V analyzed by western blotting. Cells transfected with empty vector instead of HA-ubiquitin serve as IP control (C). Actin serves as loading control. Arrow indicates IgG. (K) BAX (blue) is regulated by VDAC2 (purple)-dependent translocation to the OMM and BCL-2 protein (e.g. BCL-xL, red)-dependent retrotranslocation (left). BAX molecules that fail to retrotranslocate in a timely manner accumulate on the OMM. In this situation, BAX activation and cyt c release is prevented by BAX ubiquitylation, targeting BAX to degradation by the proteasome (yellow). Data represent mean±s.e.m. (n=3, unless otherwise indicated). *P≤0.05, one-way ANOVA. Scale bars: 10 μm.

Fig. 3.

Proteasomal degradation prevents activation of misregulated BAX. (A) BAX (blue) translocation to the mitochondria targets VDAC2 (purple) in the outer mitochondrial membrane (OMM) and VDAC2 serves as platform for BAX retrotranslocation (Lauterwasser et al., 2016). BAX is retrotranslocated upon transient interactions with BCL-xL (red). Mitochondrial association of BAX is impaired by the α5 to α6 tether (bottom left). Addition of the S184V substitution increases BAX association with VDAC2 (bottom right). (B) BN-PAGE analysis of isolated mitochondria from HCT116 BAX–BAK DKO cells expressing wild-type BAX (WT), BAX 5-6, BAX 5-6 S184V or BAX S184V using anti-VDAC or anti-BAX antibodies. Note that selected WT and 5-6 sample lanes from these blots are duplicated in Fig. 1E. (C) Confocal images of DKO cells expressing BAX S184V or BAX 5-6 S184V. Mitochondrial BAX appears yellow in the merge images. (D–G) Protein levels of BAX 5-6 S184V (D,E) and WT BAX (F,G) in the absence and the presence of the proteasome inhibitor MG132 or the ubiquitin-activating enzyme E1 enzyme inhibitor PYR41 analyzed by western blotting. VDAC serves as loading control. (H) FLIP of BAX S184V (top) or BAX 5-6 S184V (bottom) in the presence of overexpressed BCL-xL diminishes GFP fluorescence in the cytoplasm of targeted cells (outlined) completely after 80 s, and GFP fluorescence is detected only on the mitochondria (arrows). (I) Retrotranslocation of BAX S184V and BAX 5-6 S184V (blue) with BCL-xL overexpression (n≥3). (J) Immunoprecipitation of HA-tagged ubiquitin (WT, wild-type; ΔK, all lysine residues substituted to arginine) from whole-cell lysates of DKO cells expressing wild-type BAX or BAX 5-6 S184V analyzed by western blotting. Cells transfected with empty vector instead of HA-ubiquitin serve as IP control (C). Actin serves as loading control. Arrow indicates IgG. (K) BAX (blue) is regulated by VDAC2 (purple)-dependent translocation to the OMM and BCL-2 protein (e.g. BCL-xL, red)-dependent retrotranslocation (left). BAX molecules that fail to retrotranslocate in a timely manner accumulate on the OMM. In this situation, BAX activation and cyt c release is prevented by BAX ubiquitylation, targeting BAX to degradation by the proteasome (yellow). Data represent mean±s.e.m. (n=3, unless otherwise indicated). *P≤0.05, one-way ANOVA. Scale bars: 10 μm.

Parkin ubiquitylates BAX on the mitochondria

The E3 ligase parkin is recruited to the OMM to induce mitophagy (Narendra et al., 2008). To investigate whether parkin could mediate mitochondrial ubiquitylation of BAX, we analyzed potential transient interactions between BAX and parkin using a biotin ligase (BirA) protein fusion approach (Huang and Jacobson, 2010). Here, we expressed a BirA–BAX fusion protein in cells to detect transient BAX-interacting proteins (Fig. 4A). Confirming the functionality of this approach, BCL-xL was specifically biotinylated following BirA–BAX expression, in line with known transient interactions between BAX and BCL-xL. In contrast, GAPDH, an abundant protein without specific interactions to BAX, remained unlabeled. Importantly, using this approach, biotinylation of parkin was also observed, demonstrating potential transient interactions between BAX and parkin (Fig. 4B). Interestingly, the pool of labeled parkin and therefore potentially the probability of BAX interaction increases with an increased pool of misregulated BAX on the mitochondria due to the α5–α6 tether. Ectopic parkin expression enhanced BAX 5-6/S184V ubiquitylation, suggesting that parkin ubiquitylates misregulated BAX on the mitochondria (Fig. 4C). VDAC ubiquitylation by parkin was not previously reported (Geisler et al., 2010), but this might depend on massive recruitment of parkin by the mitochondria. Strikingly, two hours longer expression of wild-type parkin eliminated BAX 5-6 S184V from human cells (Fig. 4D and Fig. S5A). BAX 5-6 S184V levels are restored in the presence of parkin when proteasomal degradation is inhibited by MG132 or ubiquitylation is inhibited by PYR41.

Fig. 4.

Parkin ubiquitylates mitochondrial BAX. (A) Transient BAX protein interactions detected by BirA fusion assay in human cells. BirA-BAX fusion expression in cells (square) labels proteins (X, yellow) in close proximity to BAX with biotin (red star) by BirA. A biotin affinity matrix (solid line) separates labeled proteins (yellow) from proteins without the biotin label (Y). (B) BirA fusion assay analyzed by western blotting using indicated antibodies shows BirA–BAX interactions with parkin, in the labeled fraction (L) compared with the input (I). BCL-xL and GAPDH serve as controls. BirA–BAX is analyzed using anti-Myc antibody. (C) Immunoprecipitation of whole-cell lysates containing HA-tagged wild-type ubiquitin or ubiquitin ΔK (all lysine residues substituted to arginine) expressed with wild-type BAX or BAX 5-6 S184V in the presence of parkin in DKO cells for 5 h. BAX 5-6/S184V expression in the presence of ubiquitin ΔK but in the absence of ectopic parkin serves as control (right lane of input and IP HA). VDAC serves as loading control. Arrow indicates IgG. (D) Protein levels of BAX 5-6 S184V expressed in DKO cells with or without ectopic parkin expression for 7 h. MG132 or PYR41 prevent proteasomal degradation of BAX. VDAC serves as loading control. n=3. (E) Wild-type BAX and BAX 5-6 S184V levels with or without 10 µM CCCP for 3 h in DKO cells expressing PINK1 without ectopic parkin expression. GAPDH serves as loading control. All results are representative of n=3 replicates.

Fig. 4.

Parkin ubiquitylates mitochondrial BAX. (A) Transient BAX protein interactions detected by BirA fusion assay in human cells. BirA-BAX fusion expression in cells (square) labels proteins (X, yellow) in close proximity to BAX with biotin (red star) by BirA. A biotin affinity matrix (solid line) separates labeled proteins (yellow) from proteins without the biotin label (Y). (B) BirA fusion assay analyzed by western blotting using indicated antibodies shows BirA–BAX interactions with parkin, in the labeled fraction (L) compared with the input (I). BCL-xL and GAPDH serve as controls. BirA–BAX is analyzed using anti-Myc antibody. (C) Immunoprecipitation of whole-cell lysates containing HA-tagged wild-type ubiquitin or ubiquitin ΔK (all lysine residues substituted to arginine) expressed with wild-type BAX or BAX 5-6 S184V in the presence of parkin in DKO cells for 5 h. BAX 5-6/S184V expression in the presence of ubiquitin ΔK but in the absence of ectopic parkin serves as control (right lane of input and IP HA). VDAC serves as loading control. Arrow indicates IgG. (D) Protein levels of BAX 5-6 S184V expressed in DKO cells with or without ectopic parkin expression for 7 h. MG132 or PYR41 prevent proteasomal degradation of BAX. VDAC serves as loading control. n=3. (E) Wild-type BAX and BAX 5-6 S184V levels with or without 10 µM CCCP for 3 h in DKO cells expressing PINK1 without ectopic parkin expression. GAPDH serves as loading control. All results are representative of n=3 replicates.

Extensive parkin-dependent ubiquitylation of mitochondrial proteins has been demonstrated to follow loss in mitochondrial membrane potential [induced by the mitochondrial uncoupler carbonyl cyanide m-chlorophenyl hydrazone (CCCP)] and subsequent activation of the kinase PINK1 (Narendra et al., 2010). PINK1 phosphorylates ubiquitin and parkin directly, causing mitochondrial parkin accumulation (Kane et al., 2014; Koyano et al., 2014). Since BAX ubiquitylation is dependent on mitochondrial BAX localization, the effect of mitochondrial parkin recruitment was tested. Induction of increased recruitment of mitochondrial parkin following CCCP treatment resulted in decreased BAX 5-6 S184V levels (Fig. 4E). Increased BAX degradation after the loss of the mitochondrial membrane potential suggests enhanced BAX ubiquitylation following massive recruitment of parkin in mitochondria. By contrast, extensive mitochondrial parkin recruitment is not required for steady-state BAX ubiquitylation, and parkin activation by phosphorylation is not influenced by the presence of BAX (Fig. S5B). This also excludes the requirement of parkin S56 phosphorylation for BAX ubiquitylation (Ordureau et al., 2014). These results show that parkin ubiquitylates BAX on the OMM and this activity does not require but is enhanced by mitochondrial parkin accumulation, suggesting enhanced BAX targeting by ectopic parkin expression follows increased mitochondrial parkin levels. BAX misregulation only slightly influences transient interactions with parkin, indicating that parkin-mediated BAX ubiquitylation is dependent on the conformation of BAX.

BAX degradation is dependent on K128 ubiquitylation

Parkin specifically targets BAX but not BAK for proteasomal degradation. The protein levels of the predominantly mitochondrial BAK are not affected by parkin in the presence of ubiquitin ΔK (Fig. 5A). BAX-specific proteasomal degradation and accelerated BAX retrotranslocation ensuring low mitochondrial BAX levels reveal the paramount importance of mitochondrial BAX inhibition for cell survival. Specific ubiquitylation of BAX by parkin could theoretically occur at all nine lysine residues present in BAX. Strikingly, lysine 128 is readily exposed in the globular BAX fold between helices α5 and α6 and the corresponding residue in BAK is glycine 146 (Fig. 5B). We reasoned that ubiquitylation of K128 could confer BAX-specific degradation and tested this hypothesis introducing the K128R substitution in BAX 5-6 S184V. Indeed, the K128R substitution reduces BAX 5-6 S184V polyubiquitylation substantially (Fig. 5C). However, the inhibition of BAX polyubiquitylation is incomplete, suggesting that other lysines in overexpressed BAX are also ubiquitylated. Importantly, the BAX variant containing K128R is completely protected against parkin-dependent proteasomal BAX degradation (Fig. 5D). Therefore, polyubiquitylation in position K128 is essential for parkin-dependent proteasomal degradation of BAX.

Fig. 5.

BAX degradation is dependent on K128 ubiquitylation. (A) Western blot analysis and protein levels of wild-type BAK expressed in the absence and the presence of wild-type ubiquitin or ubiquitin ΔK (all lysine residues substituted to arginine) in DKO. VDAC serves as loading control. (B) Superimposition of the three-dimensional structures of BAX (PDB, 1F16, green) and BAK (PDB, 2IMS, cyan) showing the C-terminus of α-helices 5 and the N-termini of α-helices 6 and the turn connecting both helices in the enlargement (dashed box) made using Pymol. BAX K128 and the corresponding BAK G146 are shown. (C) Immunoprecipitation of HA-tagged ubiquitin ΔK expressed with BAX 5-6 S184 V or BAX 5-6 K128R S184V in the presence of parkin in HCT116 BAX/BAK DKO cells analyzed by western blotting. Actin serves as loading control. (D) Protein levels of BAX 5-6 S184V or BAX 5-6 K128R S184V expressed in the absence and the presence of wild-type parkin in DKO cells. VDAC serves as loading control. All results are representative of n=3 replicates.

Fig. 5.

BAX degradation is dependent on K128 ubiquitylation. (A) Western blot analysis and protein levels of wild-type BAK expressed in the absence and the presence of wild-type ubiquitin or ubiquitin ΔK (all lysine residues substituted to arginine) in DKO. VDAC serves as loading control. (B) Superimposition of the three-dimensional structures of BAX (PDB, 1F16, green) and BAK (PDB, 2IMS, cyan) showing the C-terminus of α-helices 5 and the N-termini of α-helices 6 and the turn connecting both helices in the enlargement (dashed box) made using Pymol. BAX K128 and the corresponding BAK G146 are shown. (C) Immunoprecipitation of HA-tagged ubiquitin ΔK expressed with BAX 5-6 S184 V or BAX 5-6 K128R S184V in the presence of parkin in HCT116 BAX/BAK DKO cells analyzed by western blotting. Actin serves as loading control. (D) Protein levels of BAX 5-6 S184V or BAX 5-6 K128R S184V expressed in the absence and the presence of wild-type parkin in DKO cells. VDAC serves as loading control. All results are representative of n=3 replicates.

Parkin-dependent BAX ubiquitylation protects cells from BAX activity

The combination of the intramolecular disulfide tether between helices α5 and α6 of BAX and the S184V substitution in the BAX TMD shows the importance of parkin-dependent BAX regulation, but also vastly increases the amount of misregulated BAX on the mitochondria. Therefore, the impact of parkin-mediated BAX degradation by the proteasome on mitochondrial apoptosis signaling was analyzed using ectopic wild-type BAX expression in HCT116 BAX–BAK DKO cells at near-endogenous protein levels (Todt et al., 2015). Parkin expression protects human cells from wild-type BAX activity in the absence or the presence of the apoptotic stimulus actinomycin D (Fig. 6A). A substantial increase in BAX activity with or without apoptotic stimulus is observed, when proteasomal degradation of BAX is blocked by MG132. Parkin-dependent BAX inhibition by proteasomal degradation is corroborated by a reduction in the release of cyt c (Fig. 6B). Stabilization of MCL-1 in the presence of MG132 should counteract BAX activity (Carroll et al., 2014), but profound BAX activation is observed after inhibition of the proteasomal degradation pathway. Ectopic parkin expression inhibits wild-type BAX activity in HCT116 BAX–BAK DKO cells with or without staurosporine treatment, whereas BAX K128R shows no reduction in activity in the presence of overexpressed parkin (Fig. 6C and Fig. S6). Analysis of the clonogenic cell survival of parkin-deficient HeLa cells and SH-SY5Y cells, containing high levels of endogenous parkin, in response to MG132 treatment showed MG132-concentration-dependent cell death in SH-SY5Y cells (Fig. 6D). In contrast, HeLa cells deficient in parkin-dependent BAX inhibition and thus requiring inhibition of BAX by other means, e.g. BAX retrotranslocation, show no toxic effect of MG132. YFP–parkin expression in HeLa cells significantly reduced the apoptotic response to a variety of chemotoxic stresses (Fig. 6E,F). Consequently, MG-132 treatment led to cell death in YFP–parkin-expressing HeLa cells (Fig. 6G). Parkin knockout in SH-SY5Y cells results in increased sensitivity to apoptotic stimuli (Fig. 6H). Therefore, parkin-dependent degradation of BAX provides additional protection against BAX activation when misregulated BAX accumulates on the OMM (Fig. 6I).

Fig. 6.

Parkin-dependent BAX ubiquitylation protects cells from BAX activity. (A,B) Presence of active BAX conformation analyzed by the monoclonal anti-BAX antibody 6A7 (A) and cyt c release (B) as a percentage of total BAX-expressing cells in DKO cells expressing wild-type BAX with or without parkin overexpression and the proteasome inhibitor MG132 in the absence or the presence of 1 µM actinomycin D (ActD). (C) PARP cleavage in DKO cells expressing wild-type BAX in the absence or the presence of parkin overexpression. Similar sample loading is controlled by actin. (D,G) Clonogenic survival of parkin-expressing SH-SY5Y cells (green) and parkin KO HeLa cells (blue) following incubation with 0, 5, 7.5, 10, 15 or 20 µM MG132 for 6 h. Colony formation was analyzed after 10–12 days. (E) Caspase-3/7 activity measurements of wild-type HeLa cells and HeLa cells stably expressing YFP–parkin with or without 1 µM actinomycin D (ActD), 1 µM daunorubicin (Dauno), 5 µM doxorubicin (Doxo) or 100 µM etoposide (Eto). Caspase activity is displayed in relative fluorescence units (RFU). (F) Western blot analysis of PARP cleavage following MOM permeabilization in wild-type HeLa cells (–) and HeLa cells stably expressing YFP–parkin with or without drugs as indicated. Similar sample loading is controlled by GAPDH. (G) Clonogenic survival of wild-type HeLa cells and HeLa cells stably expressing YFP–parkin following incubation with 0, 5, 7.5, 10, 15 or 20 µM MG132 for 6 h. (H) Clonogenic survival of wild-type SH-SY5Y cells (green) and SH-SY5Y parkin KO cells with or without drugs as indicated (at concentrations given in E). Colony formation was analyzed after 10–12 days. (I) BAX is inhibited by either retrotranslocation from the mitochondria into the cytosol (top) or by targeting of mitochondrial BAX to proteasomal degradation (bottom). In the presence of high levels of free pro-survival BCL-2 proteins (e.g. BCL-xL, red) BAX (blue) is regulated by VDAC2 (purple)-dependent translocation to the OMM and BCL-2 protein-dependent retrotranslocation back into the cytosol (top). Pro-survival BCL-2 proteins are thought to counteract mitochondrial parkin (green) translocation (Hollville et al., 2014) and thus parkin-dependent BAX ubiquitylation plays a minor role under these conditions. Increased stress signaling (e.g. via BH3-only proteins) reduces BAX retrotranslocation (bottom), but reduced pro-survival BCL-2 protein activities no longer prevent parkin translocation to the mitochondria. Mitochondrial parkin, in turn, targets mitochondrial BAX for degradation by the proteasome (yellow). Therefore parkin provides additional protection from mitochondrial BAX. Data represent mean±s.e.m. (n=3). *P≤0.05, ***P≤0.001, one-way ANOVA using the Holm–Sidak method.

Fig. 6.

Parkin-dependent BAX ubiquitylation protects cells from BAX activity. (A,B) Presence of active BAX conformation analyzed by the monoclonal anti-BAX antibody 6A7 (A) and cyt c release (B) as a percentage of total BAX-expressing cells in DKO cells expressing wild-type BAX with or without parkin overexpression and the proteasome inhibitor MG132 in the absence or the presence of 1 µM actinomycin D (ActD). (C) PARP cleavage in DKO cells expressing wild-type BAX in the absence or the presence of parkin overexpression. Similar sample loading is controlled by actin. (D,G) Clonogenic survival of parkin-expressing SH-SY5Y cells (green) and parkin KO HeLa cells (blue) following incubation with 0, 5, 7.5, 10, 15 or 20 µM MG132 for 6 h. Colony formation was analyzed after 10–12 days. (E) Caspase-3/7 activity measurements of wild-type HeLa cells and HeLa cells stably expressing YFP–parkin with or without 1 µM actinomycin D (ActD), 1 µM daunorubicin (Dauno), 5 µM doxorubicin (Doxo) or 100 µM etoposide (Eto). Caspase activity is displayed in relative fluorescence units (RFU). (F) Western blot analysis of PARP cleavage following MOM permeabilization in wild-type HeLa cells (–) and HeLa cells stably expressing YFP–parkin with or without drugs as indicated. Similar sample loading is controlled by GAPDH. (G) Clonogenic survival of wild-type HeLa cells and HeLa cells stably expressing YFP–parkin following incubation with 0, 5, 7.5, 10, 15 or 20 µM MG132 for 6 h. (H) Clonogenic survival of wild-type SH-SY5Y cells (green) and SH-SY5Y parkin KO cells with or without drugs as indicated (at concentrations given in E). Colony formation was analyzed after 10–12 days. (I) BAX is inhibited by either retrotranslocation from the mitochondria into the cytosol (top) or by targeting of mitochondrial BAX to proteasomal degradation (bottom). In the presence of high levels of free pro-survival BCL-2 proteins (e.g. BCL-xL, red) BAX (blue) is regulated by VDAC2 (purple)-dependent translocation to the OMM and BCL-2 protein-dependent retrotranslocation back into the cytosol (top). Pro-survival BCL-2 proteins are thought to counteract mitochondrial parkin (green) translocation (Hollville et al., 2014) and thus parkin-dependent BAX ubiquitylation plays a minor role under these conditions. Increased stress signaling (e.g. via BH3-only proteins) reduces BAX retrotranslocation (bottom), but reduced pro-survival BCL-2 protein activities no longer prevent parkin translocation to the mitochondria. Mitochondrial parkin, in turn, targets mitochondrial BAX for degradation by the proteasome (yellow). Therefore parkin provides additional protection from mitochondrial BAX. Data represent mean±s.e.m. (n=3). *P≤0.05, ***P≤0.001, one-way ANOVA using the Holm–Sidak method.

The pro-apoptotic BCL-2 protein BAX is regulated by constant translocation to the mitochondria and retrotranslocation back into the cytosol (Edlich, 2015; Edlich et al., 2011; Schellenberg et al., 2013; Todt et al., 2015). Therefore, pro-survival BCL-2 proteins inhibit BAX (and BAK) in cells by transient interactions during retrotranslocation from the OMM (Lauterwasser et al., 2016), whereas previously described heterodimeric complexes between BAX (and BAK) and pro-survival BCL-2 proteins were probably induced artificially because of the use of detergents (Hsu and Youle, 1997). BAX regulation requires major conformational changes, such as exposure of the C-terminal TMD and the BH3 domain from the globular fold (Edlich et al., 2011; Suzuki et al., 2000). Here, we show that conformational changes involving helices α5 and α6 are essential for BAX association with VDAC2 on the OMM, but also for BAX regulation by retrotranslocation into the cytosol. BAX variants suggest a central role of helix α6 in the formation of VDAC2-containing complexes on the OMM. These results support the importance of the separation of the ‘latch’ from the ‘core’ domain in BAX regulation (Czabotar et al., 2013), but also argue for a role of this conformational change in the mitochondrial association of BAX in healthy cells rather than it being solely a BAX-activation-specific event. Noteworthy, the mitochondrial BAX pool prior to apoptotic stress determines the cellular predisposition to apoptosis (Reichenbach et al., 2017; Todt et al., 2013). Therefore, the large impact of the BAX α5–α6 tether on mitochondrial BAX association is consistent with BAX activation models suggesting separation between helices α5 and α6 (Bleicken et al., 2014; Czabotar et al., 2013; Mandal et al., 2016). However, BAX 5-6 commits human cells to apoptosis and even increases apoptotic activity compared with the wild-type protein. These results suggest that BAX activation could alternatively circumvent major conformational changes between helices α5 and α6, as previously suggested (Annis et al., 2005). Therefore, BAX molecules that associate with the OMM but fail to undergo conformational changes required for BAX retrotranslocation impose the threat of OMM permeabilization upon the cell.

Proteasomal degradation of mitochondrial BAX represents an additional layer of cell protection from BAX activity. Experiments analyzing transient BAX interactions with the E3 ligase parkin suggest the importance of major conformational changes in BAX for parkin-dependent ubiquitylation. Parkin inhibits only a minor pool of endogenous BAX on the mitochondria, but in doing so provides considerable protection from BAX activity. Proteasomal degradation of BAX is induced by parkin-dependent ubiquitylation of BAX K128. Parkin has been suggested to ubiquitylate cytosolic BAX at K21 and/or K64 (Charan et al., 2014; Johnson et al., 2012). However, we do not find any support for these suggestions, as BAX ubiquitylation correlates with mitochondrial localization and BAX degradation is completely abolished by the K128R substitution. Parkin-dependent ubiquitylation regulates BAX but not the largely mitochondrial BAK, further emphasizing the importance of inhibiting BAX on the mitochondria. Parkin promotes mitophagy following selective recruitment to the OMM after loss of mitochondrial membrane potential (Narendra et al., 2008). Although mitochondrial parkin accumulation is not required for BAX ubiquitylation, loss of mitochondrial membrane potential induced by CCCP treatment increases BAX degradation. In addition, ectopic parkin expression enhances BAX ubiquitylation and degradation, depending on the ability of parkin to ubiquitylate mitochondrial proteins. Therefore, the size of the mitochondrial parkin pool determines ubiquitylation of misregulated BAX on the mitochondria.

Pro-survival BCL-2 proteins have been found to antagonize parkin translocation to the mitochondria (Hollville et al., 2014). This finding seems counterintuitive to the cell-protective function suggested for parkin in neuronal cells. Mutations in the parkin-encoding gene PRKN (also known as PARK2) occur in Parkinson's disease (PD), a condition associated with the death of dopaminergic neurons in the midbrain (Kitada et al., 1998). In addition, several studies have shown parkin-dependent cell protection from induced apoptosis (Ekholm-Reed et al., 2013; Sun et al., 2016; Wang et al., 2013). However, pro-survival BCL-2 protein-dependent inhibition of parkin translocation to the mitochondria does not contradict a central role of parkin in neuroprotection, considering the regulation of BAX. Efficient BAX retrotranslocation is ensured by high levels of pro-survival BCL-2 proteins, rendering additional inhibition of mitochondrial BAX less important (Fig. 6E). Mitochondrial parkin levels are increased when pro-survival BCL-2 proteins are inhibited and mitochondrial parkin can take over BAX inhibition on the OMM. This scenario suggests that parkin-dependent BAX ubiquitylation is a quality control mechanism balanced with BAX retrotranslocation. Pro-survival BCL-2 protein-dependent inhibition of apoptosis induced by mitochondrial depolarization further supports this connection (Carroll et al., 2014). Mitochondrial BAX inhibited by either BCL-2 protein-dependent retrotranslocation or parkin-dependent proteasomal degradation could exist in different conformations and may become active through different paths. It is possible that different activation pathways lead to different BAX complexes associated with BAX activity (Große et al., 2016; Nechushtan et al., 2001; Salvador-Gallego et al., 2016).

The bifunctional role of mitochondrial parkin in inhibiting BAX and promoting mitophagy could shape neuronal cell survival regulation. Increasing the cellular tolerance for stress could be beneficial for removal of post-mitotic neurons and thus their cell contacts could define their role. Mitochondrial parkin inhibits BAX-dependent apoptosis and induces mitophagy of impaired mitochondria, increasing stress resistance and providing the means of stress reduction at the same time. Therefore, dopaminergic neurons of patients bearing parkin mutations may undergo apoptosis that is dependent on misregulated BAX. Understanding the inhibition of mitochondrial BAX by parkin-dependent ubiquitylation and proteasomal degradation has the imminent potential to provide novel approaches for the prevention and therapy of Parkinson's disease.

Cell culture and transfection

HCT116 cells and HCT116 BAX–BAK DKO cells with or without stable GFP–BAX expression were cultured in McCoy's 5A medium supplemented with 10% heat-inactivated fetal bovine serum and 10 mM HEPES in 5% CO2 at 37°C. SH-SY5Y cells were cultured in a 1:1 mixture of DMEM and F12 medium supplemented with 10% fetal bovine serum and 10 mM HEPES in 5% CO2 at 37°C. HeLa cells were cultured in DMEM supplemented with 10% heat-inactivated fetal bovine serum and 10 mM HEPES in 5% CO2 at 37°C. Cells were transfected with Turbofect (Fermentas) or Lipofectamine LTX (Invitrogen), typically with 100 ng of BAX or BCL-xL constructs in pEGFP vector according to the manufacturer's instructions.

Maleimide–PEG (Mal–PEG) labelling

BAX variants were transiently expressed in HCT116 BAX–BAK DKO cells. Cells were lysed and the cytosolic fractions were divided into two groups for non-denaturing and denaturing conditions, adding 0.5 mM DTT and 2 mM or 0.5 mM Mal–PEG with and without 8 M urea, respectively. Samples were incubated for 1 h on ice (protected from light), subjected to acetone precipitation and analyzed by SDS-PAGE and western blotting.

Mobility shift detection of phosphorylated proteins

Parkin was expressed with and without BAX 5-6 S184 V in HCT116 BAX–BAK DKO cells for 4 h. Cells were treated with 10 µM CCCP for 3 h or left untreated. Membrane fractions were prepared from transfected cells. For the detection of protein phosphorylation, an acrylamide based Phos-tag gel (4.5%, Phos-tag and MnCl2, without EDTA) was used according to the manufacturer's protocol (Wako Chemicals).

Whole-cell lysis, subcellular fractionation

Cells were harvested and incubated in cell lysis buffer [10 mM HEPES, pH 7.4, 150 mM NaCl, 1% Triton X-100, protease inhibitor cocktail (Roche)] for 15 min on ice. Whole-cell extracts were obtained by centrifugation at 15,000 g for 10 min at 4°C. Samples were boiled in SDS sample buffer for 5 min at 95°C and subsequently subjected to SDS-PAGE and western blot analysis. Subcellular fractionations were performed as previously described (Todt et al., 2013).

Confocal microscopy and FLIP

HCT116 BAX–BAK DKO GFP–BAX-expressing cells were seeded on a chambered cover glass (Thermo Fisher Scientific) in McCoy's 5A medium, grown for 36 h and imaged using a Zeiss 510 META confocal LSM microscope equipped with argon (458/488/514 nm lines) and HeNe (543/633 nm) lasers.

Fluorescence loss in photobleaching (FLIP) experiments were performed as described previously (Edlich et al., 2011). In short, cells were imaged prior to bleaching then a single region (diameter of 1 μm) within the nucleus was repeatedly bleached with two iterations of a 488 nm laser line (100% output) using a Zeiss LSM510 META with a 63× PlanFluor lens. Two images were collected after each bleach pulse. After 15 cycles of bleaching and collecting 30 images, separate measurements on the mitochondria were taken to analyze loss of fluorescence. Unbleached cells neighboring analyzed cells served as controls for photobleaching during image acquisition of each measurement. Fluorescence intensities were normalized by setting the pre-bleach fluorescence to 100% signal.

For 6A7 staining and cyt c release, BAX variants and parkin constructs were transfected into HCT116 BAX–BAK DKO cells and then, cells were treated with either 20 µM Mg132 for 3 h or left as control; 1 µM ActD was used as apoptotic stimulus. After the treatment, cells were fixed with 4% paraformaldehyde and permeabilized with Triton X-100 (0.15%). Anti-BAX 6A7 antibody (Sigma, 1:500) or anti-cyt c antibody (6H2B4, BD Biosciences, 1:1000) were added for 1 h at room temperature (RT). After washing with PBS, cells were incubated with the secondary antibodies mouse Alexa Fluor 594 (Invitrogen, 1:750) and rabbit Alexa Fluor 647 (Invitrogen, 1:1000), for 1 h at RT. Cells were washed with PBS and GFP-positive cells were quantified.

Apoptosis activity assays

Apoptosis was induced using STS (Staurosporin, Enzo Life Technologies) or ActD (ActinomycinD, Sigma Aldrich). For caspase assays, cells were washed with ice-cold 1×PBS and resuspended in ice-cold cell lysis buffer (20 mM Tris-HCl, pH 7.4, 100 mM NaCl, 1 mM EDTA, with protease inhibitor cocktail and 0.5% Triton X-100). Whole-cell lysate was incubated with caspase-3/7 substrate (BD Pharmingen) for 60 min at 37°C and protein concentration was determined by a Bradford Assay (Roth). Substrate cleavage was measured for 50 cycles with 10 s delay (excitation at 380 nm, emission at 430–460 nm). Kinetics were determined and calculated to the amount of protein per sample.

Analysis of native protein complexes

To analyze native protein complexes, isolated mitochondria were solubilized in digitonin buffer (20 mM Tris-HCl, pH 7.4, 50 mM NaCl, 0.1 mM EDTA, 10% v/v glycerol, 2% w/v digitonin, 1 mM PMSF) and subsequently incubated at 4°C for 45 min in an end-over-end shaker. After insoluble material was removed by centrifugation (16,000 g, 10 min, 4°C) mitochondrial extracts were subjected to BN-PAGE (Schägger and von Jagow, 1991) using 4–13% continuous polyacrylamide gradient gels.

Immunoprecipitation of ubiquitylated proteins

HCT116 BAX–BAK DKO cells transfected with HA-tagged ubiquitin, GFP-BAX and YFP-parkin as indicated, were harvested in ice-cold PBS and lysed in IP buffer [10 mM HEPES, pH 7.4, 150 mM NaCl, containing complete protease inhibitor mix (Roche) and 0.2% Triton X-100]. After clearing the lysate by centrifugation at 15,000 g for 10 min, input sample (1.25%) was separated. The remaining lysate was incubated with HA-antibody coupled agarose beads (Santa Cruz), end-over-end shaking at 4°C overnight. After incubation, the beads were washed five times with IP buffer and finally boiled in SDS loading buffer. Input and bead samples were resolved by 10% SDS-PAGE and analyzed by western blotting for the indicated proteins. Antibodies used were: GFP D5.1 (Cell Signaling), Myc 9B11 (Cell Signaling), AKT1 2H10 (Cell Signaling), VDAC/Porin ab5 (Calbiochem), VDAC N-18 (Santa Cruz), PARP (Cell Signaling), Actin C4 (Millipore), BAX E63 (Abcam), TOM20 FL-145 (Santa Cruz), HA Y11 (Santa Cruz), Bcl XL 54H6 (Cell Signaling), Parkin PRK8 (Santa Cruz), GAPDH (Sigma), BAK (Millipore), K48 Polyubiquitin (Cell Signaling) and K63 Polyubiquitin (Cell Signaling). pRK5-HA-ubiquitin-KO and pRK5-HA-ubiquitin-WT were obtained from AddGene and were deposited by Ted Dawson [plasmids #17603 and #17608 (Lim et al., 2005)].

BirA assay

HCT116 BAX–BAK DKO cells were transfected with pcDNA3–mycBioID–BAX plasmids, resulting in Myc-tagged BirA–BAX fusion expression. After cell harvest in ice-cold PBS, the cell pellet was resuspended and lysed in IP buffer [10 mM HEPES, pH 7.4, 150 mM NaCl, complete proteinase inhibitor cocktail (Roche) and 0.2% Triton X-100]. Cell lysate was centrifuged (120,000 g, 30 min, 4°C) and washed using a gel filtration column (GE Healthcare). Then, input sample (2.5%) was separated and the remaining lysate incubated with streptavidin agarose beads (Thermo, overnight at 4°C). Subsequently, beads were washed and boiled in SDS sample buffer. Input and pull-down samples were analyzed by SDS-PAGE and western blotting.

Clonogenic survival assay

HeLa and SHSY-5Y cells were seeded to 70% confluency, treated for 6 h with 0, 5, 7.5, 10, 15 or 20 µM MG132 in DMSO. Cells were cultured for 10–12 days followed by fixation with 4% PFA in PBS and staining with Methylene Blue.

We thank R. Youle for providing YFP–parkin-expressing HeLa cells and X. Luo for sharing BAX ΔH6 with us.

Author contributions

Conceptualization: F.E.; Methodology: Z.C., J.L., F.T., K.F., R.M.Z., A.Ö.; Validation: Z.C., J.L., A.T., M.v.d.L.; Writing - original draft: Z.C., F.E.; Writing - review & editing: A.T., M.v.d.L., F.E.; Visualization: F.E.; Supervision: F.E.; Project administration: F.E.; Funding acquisition: F.E.

Funding

This work is supported by the Emmy Noether program, the Heisenberg program and the Sonderforschungsbereich 746 of the German Research Council (Deutsche Forschungsgemeinschaft, DFG), the Else Kröner-Fresenius-Stiftung, the Wilhelm Sander-Stiftung, the Excellence Initiative of the DFG (Spemann Graduate School, GSC-4), the Centre for Biological Signalling Studies (BIOSS, EXC-294) funded by the Bundesministerium für Bildung und Forschung. A.T. was supported by The Cell Science Research Foundation and JSPS KAKENHI (JP24111513, JP26870067, JP16K19047).

Annis
,
M. G.
,
Soucie
,
E. L.
,
Dlugosz
,
P. J.
,
Cruz-Aguado
,
J. A.
,
Penn
,
L. Z.
,
Leber
,
B.
and
Andrews
,
D. W.
(
2005
).
Bax forms multispanning monomers that oligomerize to permeabilize membranes during apoptosis
.
EMBO J.
24
,
2096
-
2103
.
Bleicken
,
S.
,
Jeschke
,
G.
,
Stegmueller
,
C.
,
Salvador-Gallego
,
R.
,
García-Sáez
,
A. J.
and
Bordignon
,
E.
(
2014
).
Structural model of active Bax at the membrane
.
Mol. Cell
56
,
496
-
505
.
Carroll
,
R. G.
,
Hollville
,
E.
and
Martin
,
S. J.
(
2014
).
Parkin sensitizes toward apoptosis induced by mitochondrial depolarization through promoting degradation of Mcl-1
.
Cell Rep.
9
,
1538
-
1553
.
Charan
,
R. A.
,
Johnson
,
B. N.
,
Zaganelli
,
S.
,
Nardozzi
,
J. D.
and
LaVoie
,
M. J.
(
2014
).
Inhibition of apoptotic Bax translocation to the mitochondria is a central function of parkin
.
Cell Death Dis.
5
,
e1313
.
Cheng
,
E. H.-Y.
,
Sheiko
,
T. V.
,
Fisher
,
J. K.
,
Craigen
,
W. J.
and
Korsmeyer
,
S. J.
(
2003
).
VDAC2 inhibits BAK activation and mitochondrial apoptosis
.
Science
301
,
513
-
517
.
Czabotar
,
P. E.
,
Westphal
,
D.
,
Dewson
,
G.
,
Ma
,
S.
,
Hockings
,
C.
,
Fairlie
,
W. D.
,
Lee
,
E. F.
,
Yao
,
S.
,
Robin
,
A. Y.
,
Smith
,
B. J.
, et al. 
(
2013
).
Bax crystal structures reveal how BH3 domains activate Bax and nucleate its oligomerization to induce apoptosis
.
Cell
152
,
519
-
531
.
Edlich
,
F.
(
2015
).
The great migration of Bax and Bak
.
Mol. Cell. Oncol.
2
,
e995029
.
Edlich
,
F.
,
Banerjee
,
S.
,
Suzuki
,
M.
,
Cleland
,
M. M.
,
Arnoult
,
D.
,
Wang
,
C.
,
Neutzner
,
A.
,
Tjandra
,
N.
and
Youle
,
R. J.
(
2011
).
Bcl-x(L) retrotranslocates Bax from the mitochondria into the cytosol
.
Cell
145
,
104
-
116
.
Ekholm-Reed
,
S.
,
Goldberg
,
M. S.
,
Schlossmacher
,
M. G.
and
Reed
,
S. I.
(
2013
).
Parkin-dependent degradation of the F-box protein Fbw7β promotes neuronal survival in response to oxidative stress by stabilizing Mcl-1
.
Mol. Cell. Biol.
33
,
3627
-
3643
.
Geisler
,
S.
,
Holmström
,
K. M.
,
Skujat
,
D.
,
Fiesel
,
F. C.
,
Rothfuss
,
O. C.
,
Kahle
,
P. J.
and
Springer
,
W.
(
2010
).
PINK1/Parkin-mediated mitophagy is dependent on VDAC1 and p62/SQSTM1
.
Nat. Cell Biol.
12
,
119
-
131
.
George
,
N. M.
,
Evans
,
J. J. D.
and
Luo
,
X.
(
2007
).
A three-helix homo-oligomerization domain containing BH3 and BH1 is responsible for the apoptotic activity of Bax
.
Genes Dev.
21
,
1937
-
1948
.
Große
,
L.
,
Wurm
,
C. A.
,
Brüser
,
C.
,
Neumann
,
D.
,
Jans
,
D. C.
and
Jakobs
,
S.
(
2016
).
Bax assembles into large ring-like structures remodeling the mitochondrial outer membrane in apoptosis
.
EMBO J.
35
,
402
-
413
.
Hollville
,
E.
,
Carroll
,
R. G.
,
Cullen
,
S. P.
and
Martin
,
S. J.
(
2014
).
Bcl-2 family proteins participate in mitochondrial quality control by regulating Parkin/PINK1-dependent mitophagy
.
Mol. Cell
55
,
451
-
466
.
Hotchkiss
,
R. S.
,
Strasser
,
A.
,
McDunn
,
J. E.
and
Swanson
,
P. E.
(
2009
).
Cell death
.
N. Engl. J. Med.
361
,
1570
-
1583
.
Hsu
,
Y.-T.
and
Youle
,
R. J.
(
1997
).
Nonionic detergents induce dimerization among members of the Bcl-2 family
.
J. Biol. Chem.
272
,
13829
-
13834
.
Hsu
,
Y.-T.
and
Youle
,
R. J.
(
1998
).
Bax in murine thymus is a soluble monomeric protein that displays differential detergent-induced conformations
.
J. Biol. Chem.
273
,
10777
-
10783
.
Huang
,
C.
and
Jacobson
,
K.
(
2010
).
Detection of protein-protein interactions using nonimmune IgG and BirA-mediated biotinylation
.
BioTechniques
49
,
881
-
886
.
Ichim
,
G.
,
Lopez
,
J.
,
Ahmed
,
S. U.
,
Muthalagu
,
N.
,
Giampazolias
,
E.
,
Delgado
,
M. E.
,
Haller
,
M.
,
Riley
,
J. S.
,
Mason
,
S. M.
,
Athineos
,
D.
, et al. 
(
2015
).
Limited mitochondrial permeabilization causes DNA damage and genomic instability in the absence of cell death
.
Mol. Cell
57
,
860
-
872
.
Johnson
,
B. N.
,
Berger
,
A. K.
,
Cortese
,
G. P.
and
Lavoie
,
M. J.
(
2012
).
The ubiquitin E3 ligase parkin regulates the proapoptotic function of Bax
.
Proc. Natl. Acad. Sci. USA
109
,
6283
-
6288
.
Kane
,
L. A.
,
Lazarou
,
M.
,
Fogel
,
A. I.
,
Li
,
Y.
,
Yamano
,
K.
,
Sarraf
,
S. A.
,
Banerjee
,
S.
and
Youle
,
R. J.
(
2014
).
PINK1 phosphorylates ubiquitin to activate Parkin E3 ubiquitin ligase activity
.
J. Cell Biol.
205
,
143
-
153
.
Kitada
,
T.
,
Asakawa
,
S.
,
Hattori
,
N.
,
Matsumine
,
H.
,
Yamamura
,
Y.
,
Minoshima
,
S.
,
Yokochi
,
M.
,
Mizuno
,
Y.
and
Shimizu
,
N.
(
1998
).
Mutations in the parkin gene cause autosomal recessive juvenile parkinsonism
.
Nature
392
,
605
-
608
.
Koyano
,
F.
,
Okatsu
,
K.
,
Kosako
,
H.
,
Tamura
,
Y.
,
Go
,
E.
,
Kimura
,
M.
,
Kimura
,
Y.
,
Tsuchiya
,
H.
,
Yoshihara
,
H.
,
Hirokawa
,
T.
, et al. 
(
2014
).
Ubiquitin is phosphorylated by PINK1 to activate parkin
.
Nature
510
,
162
-
166
.
Lauterwasser
,
J.
,
Todt
,
F.
,
Zerbes
,
R. M.
,
Nguyen
,
T. N.
,
Craigen
,
W.
,
Lazarou
,
M.
,
van der Laan
,
M.
and
Edlich
,
F.
(
2016
).
The porin VDAC2 is the mitochondrial platform for Bax retrotranslocation
.
Sci. Rep.
6
,
32994
.
Lazarou
,
M.
,
Stojanovski
,
D.
,
Frazier
,
A. E.
,
Kotevski
,
A.
,
Dewson
,
G.
,
Craigen
,
W. J.
,
Kluck
,
R. M.
,
Vaux
,
D. L.
and
Ryan
,
M. T.
(
2010
).
Inhibition of Bak activation by VDAC2 is dependent on the Bak transmembrane anchor
.
J. Biol. Chem.
285
,
36876
-
36883
.
Letai
,
A.
,
Bassik
,
M. C.
,
Walensky
,
L. D.
,
Sorcinelli
,
M. D.
,
Weiler
,
S.
and
Korsmeyer
,
S. J.
(
2002
).
Distinct BH3 domains either sensitize or activate mitochondrial apoptosis, serving as prototype cancer therapeutics
.
Cancer Cell
2
,
183
-
192
.
Lim
,
K. L.
,
Chew
,
K. C.
,
Tan
,
J. M.
,
Wang
,
C.
,
Chung
,
K. K.
,
Zhang
,
Y.
,
Tanaka
,
Y.
,
Smith
,
W.
,
Engelender
,
S.
, et al. 
(
2005
).
Parkin mediates nonclassical, proteasomal-independent ubiquitination of synphilin-1: implications for Lewy body formation
.
J. Neurosci
.
25
,
2002
-
2009
.
Llambi
,
F.
,
Moldoveanu
,
T.
,
Tait
,
S. W. G.
,
Bouchier-Hayes
,
L.
,
Temirov
,
J.
,
McCormick
,
L. L.
,
Dillon
,
C. P.
and
Green
,
D. R.
(
2011
).
A Unified Model of Mammalian BCL-2 Protein Family Interactions at the Mitochondria
.
Mol. Cell
44
,
517
-
531
.
Ma
,
S. B.
,
Nguyen
,
T. N.
,
Tan
,
I.
,
Ninnis
,
R.
,
Iyer
,
S.
,
Stroud
,
D. A.
,
Menard
,
M.
,
Kluck
,
R. M.
,
Ryan
,
M. T.
and
Dewson
,
G.
(
2014
).
Bax targets mitochondria by distinct mechanisms before or during apoptotic cell death: a requirement for VDAC2 or Bak for efficient Bax apoptotic function
.
Cell Death Differ.
21
,
1925
-
1935
.
Mandal
,
T.
,
Shin
,
S.
,
Aluvila
,
S.
,
Chen
,
H.-C.
,
Grieve
,
C.
,
Choe
,
J.-Y.
,
Cheng
,
E. H.
,
Hustedt
,
E. J.
and
Oh
,
K. J.
(
2016
).
Assembly of Bak homodimers into higher order homooligomers in the mitochondrial apoptotic pore
.
Sci. Rep.
6
,
30763
.
Martinou
,
J.-C.
and
Youle
,
R. J.
(
2011
).
Mitochondria in apoptosis: Bcl-2 family members and mitochondrial dynamics
.
Dev. Cell
21
,
92
-
101
.
Narendra
,
D.
,
Tanaka
,
A.
,
Suen
,
D.-F.
and
Youle
,
R. J.
(
2008
).
Parkin is recruited selectively to impaired mitochondria and promotes their autophagy
.
J. Cell Biol.
183
,
795
-
803
.
Narendra
,
D. P.
,
Jin
,
S. M.
,
Tanaka
,
A.
,
Suen
,
D.-F.
,
Gautier
,
C. A.
,
Shen
,
J.
,
Cookson
,
M. R.
and
Youle
,
R. J.
(
2010
).
PINK1 is selectively stabilized on impaired mitochondria to activate Parkin
.
PLoS Biol.
8
,
e1000298
.
Nechushtan
,
A.
,
Smith
,
C. L.
,
Hsu
,
Y. T.
and
Youle
,
R. J.
(
1999
).
Conformation of the Bax C-terminus regulates subcellular location and cell death
.
EMBO J.
18
,
2330
-
2341
.
Nechushtan
,
A.
,
Smith
,
C. L.
,
Lamensdorf
,
I.
,
Yoon
,
S.-H.
and
Youle
,
R. J.
(
2001
).
Bax and Bak coalesce into novel mitochondria-associated clusters during apoptosis
.
J. Cell Biol.
153
,
1265
-
1276
.
Ordureau
,
A.
,
Sarraf
,
S. A.
,
Duda
,
D. M.
,
Heo
,
J.-M.
,
Jedrychowski
,
M. P.
,
Sviderskiy
,
V. O.
,
Olszewski
,
J. L.
,
Koerber
,
J. T.
,
Xie
,
T.
,
Beausoleil
,
S. A.
, et al. 
(
2014
).
Quantitative proteomics reveal a feedforward mechanism for mitochondrial PARKIN translocation and ubiquitin chain synthesis
.
Mol. Cell
56
,
360
-
375
.
Reichenbach
,
F.
,
Wiedenmann
,
C.
,
Schalk
,
E.
,
Becker
,
D.
,
Funk
,
K.
,
Scholz-Kreisel
,
P.
,
Todt
,
F.
,
Wolleschak
,
D.
,
Dohner
,
K.
,
Marquardt
,
J. U.
, et al. 
(
2017
).
Mitochondrial BAX determines the predisposition to apoptosis in human AML
.
Clin. Cancer Res.
doi:10.1158/1078-0432.CCR-16-1941 [Epub ahead of print]
.
Salvador-Gallego
,
R.
,
Mund
,
M.
,
Cosentino
,
K.
,
Schneider
,
J.
,
Unsay
,
J.
,
Schraermeyer
,
U.
,
Engelhardt
,
J.
,
Ries
,
J.
and
García-Sáez
,
A. J.
(
2016
).
Bax assembly into rings and arcs in apoptotic mitochondria is linked to membrane pores
.
EMBO J.
35
,
389
-
401
.
Samuel
,
T.
,
Weber
,
H. O.
,
Rauch
,
P.
,
Verdoodt
,
B.
,
Eppel
,
J.-T.
,
McShea
,
A.
,
Hermeking
,
H.
and
Funk
,
J. O.
(
2001
).
The G2/M regulator 14-3-3sigma prevents apoptosis through sequestration of Bax
.
J. Biol. Chem.
276
,
45201
-
45206
.
Schägger
,
H.
and
von Jagow
,
G.
(
1991
).
Blue native electrophoresis for isolation of membrane protein complexes in enzymatically active form
.
Anal. Biochem.
199
,
223
-
231
.
Schellenberg
,
B.
,
Wang
,
P.
,
Keeble
,
J. A.
,
Rodriguez-Enriquez
,
R.
,
Walker
,
S.
,
Owens
,
T. W.
,
Foster
,
F.
,
Tanianis-Hughes
,
J.
,
Brennan
,
K.
,
Streuli
,
C. H.
, et al. 
(
2013
).
Bax exists in a dynamic equilibrium between the cytosol and mitochondria to control apoptotic priming
.
Mol. Cell
49
,
959
-
971
.
Shen
,
Z.-J.
,
Esnault
,
S.
,
Schinzel
,
A.
,
Borner
,
C.
and
Malter
,
J. S.
(
2009
).
The peptidyl-prolyl isomerase Pin1 facilitates cytokine-induced survival of eosinophils by suppressing Bax activation
.
Nat. Immunol.
10
,
257
-
265
.
Sun
,
L.
,
Zhao
,
M.
,
Yang
,
Y.
,
Xue
,
R.-Q.
,
Yu
,
X.-J.
,
Liu
,
J.-K.
and
Zang
,
W.-J.
(
2016
).
Acetylcholine Attenuates Hypoxia/Reoxygenation Injury by Inducing Mitophagy Through PINK1/Parkin Signal Pathway in H9c2 Cells
.
J. Cell. Physiol.
231
,
1171
-
1181
.
Suzuki
,
M.
,
Youle
,
R. J.
and
Tjandra
,
N.
(
2000
).
Structure of Bax: coregulation of dimer formation and intracellular localization
.
Cell
103
,
645
-
654
.
Suzuki
,
M.
,
Jeong
,
S.-Y.
,
Karbowski
,
M.
,
Youle
,
R. J.
and
Tjandra
,
N.
(
2003
).
The solution structure of human mitochondria fission protein Fis1 reveals a novel TPR-like helix bundle
.
J. Mol. Biol.
334
,
445
-
458
.
Tait
,
S. W. G.
and
Green
,
D. R.
(
2010
).
Mitochondria and cell death: outer membrane permeabilization and beyond
.
Nat. Rev. Mol. Cell Biol.
11
,
621
-
632
.
Todt
,
F.
,
Cakir
,
Z.
,
Reichenbach
,
F.
,
Youle
,
R. J.
and
Edlich
,
F.
(
2013
).
The C-terminal helix of Bcl-x(L) mediates Bax retrotranslocation from the mitochondria
.
Cell Death Differ.
20
,
333
-
342
.
Todt
,
F.
,
Cakir
,
Z.
,
Reichenbach
,
F.
,
Emschermann
,
F.
,
Lauterwasser
,
J.
,
Kaiser
,
A.
,
Ichim
,
G.
,
Tait
,
S. W. G.
,
Frank
,
S.
,
Langer
,
H. F.
, et al. 
(
2015
).
Differential retrotranslocation of mitochondrial Bax and Bak
.
EMBO J.
34
,
67
-
80
.
Tsuruta
,
F.
,
Sunayama
,
J.
,
Mori
,
Y.
,
Hattori
,
S.
,
Shimizu
,
S.
,
Tsujimoto
,
Y.
,
Yoshioka
,
K.
,
Masuyama
,
N.
and
Gotoh
,
Y.
(
2004
).
JNK promotes Bax translocation to mitochondria through phosphorylation of 14-3-3 proteins
.
EMBO J.
23
,
1889
-
1899
.
Vogel
,
S.
,
Raulf
,
N.
,
Bregenhorn
,
S.
,
Biniossek
,
M. L.
,
Maurer
,
U.
,
Czabotar
,
P.
and
Borner
,
C.
(
2012
).
Cytosolic Bax: does it require binding proteins to keep its pro-apoptotic activity in check?
J. Biol. Chem.
287
,
9112
-
9127
.
Wang
,
C.
and
Youle
,
R. J.
(
2012
).
Predominant requirement of Bax for apoptosis in HCT116 cells is determined by Mcl-1's inhibitory effect on Bak
.
Oncogene
31
,
3177
-
3189
.
Wang
,
D. B.
,
Garden
,
G. A.
,
Kinoshita
,
C.
,
Wyles
,
C.
,
Babazadeh
,
N.
,
Sopher
,
B.
,
Kinoshita
,
Y.
and
Morrison
,
R. S.
(
2013
).
Declines in Drp1 and parkin expression underlie DNA damage-induced changes in mitochondrial length and neuronal death
.
J. Neurosci. Off. J. Soc. Neurosci.
33
,
1357
-
1365
.
Willis
,
S. N.
,
Chen
,
L.
,
Dewson
,
G.
,
Wei
,
A.
,
Naik
,
E.
,
Fletcher
,
J. I.
,
Adams
,
J. M.
and
Huang
,
D. C. S.
(
2005
).
Proapoptotic Bak is sequestered by Mcl-1 and Bcl-xL, but not Bcl-2, until displaced by BH3-only proteins
.
Genes Dev.
19
,
1294
-
1305
.
Willis
,
S. N.
,
Fletcher
,
J. I.
,
Kaufmann
,
T.
,
van Delft
,
M. F.
,
Chen
,
L.
,
Czabotar
,
P. E.
,
Ierino
,
H.
,
Lee
,
E. F.
,
Fairlie
,
W. D.
,
Bouillet
,
P.
, et al. 
(
2007
).
Apoptosis initiated when BH3 ligands engage multiple Bcl-2 homologs, not Bax or Bak
.
Science
315
,
856
-
859
.
Youle
,
R. J.
and
Strasser
,
A.
(
2008
).
The BCL-2 protein family: opposing activities that mediate cell death
.
Nat. Rev. Mol. Cell Biol.
9
,
47
-
59
.

Competing interests

The authors declare no competing or financial interests.

Supplementary information