Abstract
The Bcl-2 family of proteins includes pro- and anti-apoptotic factors acting at mitochondrial and microsomal membranes. An impressive body of published studies, using genetic and physical reconstitution experiments in model organisms and cell lines, supports a view of Bcl-2 proteins as the critical arbiters of apoptotic cell death decisions in most circumstances (excepting CD95 death receptor signaling in Type I cells). Evasion of apoptosis is one of the hallmarks of cancer1 (Hanahan and Weinberg), relevant to tumorigenesis as well as resistance to cytotoxic drugs, and deregulation of Bcl-2 proteins is observed in many cancers2,3 (Manion 2003, Olejniczak 2007). The rekindled interest in aerobic glycolysis as a cancer trait raises interesting questions as to how metabolic changes in cancer cells are integrated with other essential alterations in cancer, e.g. promotion of angiogenesis and unbridled growth signals. Apoptosis induced by multiple different signals involves loss of mitochondrial homeostasis, in particular, outer mitochondrial membrane integrity, releasing cytochrome c and other proteins from the intermembrane space. This integrative process, controlled by Bcl-2 family proteins, is also influenced by the metabolic state of the cell. In this review, we consider the role of reactive oxygen species, a metabolic by-product, in the mitochondrial pathway of apoptosis, and the relationships between Bcl-2 functions and oxidative stress.
Keywords: Apoptosis, mitochondria, reactive oxygen species
The extended Bcl-2 family
The discovery of BCL2 in 1985 as the oncogene harbored in t(14;18) translocations pathognomonic of follicular B-cell lymphomas was defining in several aspects. BCL2 became the first gene identified as an apoptosis regulator, soon bolstered by the reporting of ced-9, the homologous inhibitor of programmed cell death in C. elegans, and helped establish apoptosis as a specific pathway of cellular demise [4,5]. Prior to BCL2, oncogenes were recognized as promoting cell proliferation and transformation. As BCL2 lacked these effects, cooperating effects of oncogenes could be rationally understood in terms of complementing functions [6]. And finally, the localization of Bcl-2 protein to mitochondria provided an initial inkling of the importance of this organelle in apoptosis [7].
More than half of the Bcl-2 protein family promote apoptosis, in opposition to Bcl-2, including the second family member identified, BAX. All anti-apoptotic members and 3 pro-apoptotic proteins (Bax, Bak and Bok) contain 3 or 4 conserved domains (BH1–BH4) and a C-terminal transmembrane segment [8]. Three-dimensional protein structures of these proteins show globular proteins with similar all-alpha helical folds, without a clear-cut distinction at a structural level between those with pro- or anti-apoptotic functions [9]. The largest class of Bcl-2 proteins have a single conserved domain, the so-called BH3-only proteins. Although the Bcl-2 fold can be retained in this class, many of these proteins appear unstructured in solution [10]. Genomic sequencing efforts have revealed Bcl-2-related genes in all metazoan phyla examined to date.
Bcl-2 locus of action
Bcl-2 family proteins are found at mitochondrial and endoplasmic reticulum sites, as soluble proteins in cytosolic fractions, or bound to cytoskeletal elements [11]. The C-terminal tail of Bcl-2 functions as a signal anchor sequence for mitochondrial import, where it interacts with the import receptor Tom20 and the FKBP38 immunophilin, or delivery to the endoplasmic reticulum [12–15]. Several proteins, including Bcl-xL, Bax, and multiple BH3-only proteins, exhibit facultative targeting, with redistribution from cytosol-to-mitochondria in response to apoptotic stimuli [16]. In the case of multi-domain proteins, the C-terminal transmembrane domain is folded into a hydrophobic groove on the protein surface, requiring displacement by a BH3-only protein or conformational change affecting an N-terminal mitochondrial addressing signal [16–18]. BH3-only proteins expressed in healthy cells are sequestered by cytosolic proteins (Bad), attached to cytoskeletal elements (Bim), or exist as soluble, globular proteins (Bid).
The sine qua non of Bcl-2 survival function is mitochondrial retention of intermembrane space proteins including cytochrome c. Mislocalized cytochrome c acts as a cofactor for assembly of apoptosome complexes activating caspase-9. Release of other mitochondrial proteins competitively displace caspases-3, 7 and 9 from XIAP (SMAC and HtrA2/Omi), and induce caspase-independent chromatin condensation and DNA fragmentation (AIF, endonuclease G). The pro-apoptotic Bax and Bak proteins are leading candidates as apoptotic protein pores as a result of membrane embedding and oligomerization [19].
The terminal conformational changes leading to pore assembly are promoted by interactions with BH3-only proteins and membrane lipids [20]. The anti-apoptotic functions of Bcl-2 proteins can be explained in this model by inhibitory associations with the pore-forming Bax and Bak proteins or activating BH3-only proteins [21]. In some cases, Bcl-2 inhibits changes in N-terminal conformation of cytosolic Bax and Bax translocation to mitochondrial sites, suggesting a mechanism of action that does not require physical association [22,23].
Competing models for cytochrome c release during apoptosis involve osmotic swelling of the mitochondrial matrix with subsequent rupture of the less-expansible outer membrane. Mitochondria isolated from growth factor-deprived cells are insensitive to respiratory control by external ADP and accumulate creatine phosphate in the intermembrane space, consistent with closure of the voltage-dependent anion channel (VDAC) in the outer mitochondrial membrane (OMM) [24]. Inner mitochondrial membrane hyperpolarization resulting from ATP/ADP exchange leads to matrix swelling. Bcl-xL expression sustains OMM permeability under these conditions, and increases the VDAC gating potential in reconstituted preparations [25]. Similar changes were not observed with Fas- or TNF-induced apoptosis, suggesting alternative Bcl-xL-inhibited mechanisms for cytochrome c release [24]. Bcl-2 also inhibits opening of mitochondrial permeability transition pores, another mechanism of matrix swelling, induced by diverse agents (atractyloside, uncoupling agents, alkylperoxides), although the relevance of mitochondrial permeability transition for apoptosis and Bcl-2 has been called into question [26–28].
Extra-mitochondrial functions of Bcl-2 can also sustain mitochondrial integrity. Activation of ER-localized Bax or Bak results in release of Ca2+ stores, which can promote cytochrome c release through ER-mitochondrial crosstalk. Bcl-2/Bcl-xL expression affects Ca2+ transport, independently of Bax/Bak, through direct interactions with the inositol 1,4,5-triphosphate receptor and sarco/ER Ca2+ -ATPase [29–31].
Oxidative stress in apoptosis
Apoptotic cell death can be triggered by a very long list of environmental conditions, pharmacologic agents, imbalanced signal transduction pathways, and minor damages to various cellular constituents. A majority of studies in the literature have shown that reducing oxidative stress prevents apoptosis, including instances without obvious pro-oxidant triggers. Examples include glucocorticoid-induced death of T-lymphocytes [32–35], hyper- and hypo-thermia [36–40], growth factor or serum deprivation [41–46], TNF, FasL, and TRAIL-engaged death receptor signaling [47–61], viral infection [62,63], c-myc- and p53-induced apoptosis [64–68] and staurosporine-induced cell death [69–72].
Counter-arguments against a role for oxidative stress in apoptosis have been raised, including the ability for apoptosis to proceed normally in hypoxic environments. Normal apoptotic responses to staurosporine, anti-Fas antibody, and IL-3 deprivation occur in cells grown in very low oxygen environments (< 8 ppm) to eliminate reactive oxygen species (ROS) formation [73,74]. Other studies have demonstrated that ROS are still produced by submitochondrial particles at low oxygen tensions, generated by near-complete oxidation of succinate and to a lesser extent, NADH [75]. Hypoxia paradoxically increases superoxide production, primarily at complex III of the electron transport chain [76]. This appears to due to the maintenance of normoxic levels of flux through the electron transport chain (ETC) at very low oxygen tensions (half-maximal O2 consumption < 45 ppm at low [ATP]/[ADP]•[Pi] ratios) [77]. Cytochrome c oxidase (complex IV) activity is reduced in hypoxia with a compensatory shift toward a reduced state of cytochrome c and more proximal electron carriers [78,79], leading to superoxide (O2−) production at complex III [80,81]. The ability to induce apoptosis under stringently anoxic conditions suggests that ROS are dispensible for apoptosis under these conditions [82], although equilibration with liquid media and oxygen absorption by plasticware may complicate interpretation of hypoxia experiments [83,84]. Surprisingly, ρ° cells lacking mitochondrial DNA and a functional electron transport chain, are resistant to anoxia-induced apoptosis. This raises the interesting possibility that mammalian cells are capable of anaerobic mitochondrial metabolism for redox balance or ATP generation during anoxia, e.g. using nitrite as a terminal electron acceptor, as occurs in plants [85,86]
Although outnumbered by positive studies, a number of studies have failed to demonstrate inhibition of apoptosis using anti-oxidants [87,88]. One caveat in interpreting these studies is the potential for anti-oxidant compounds to exhibit pro-oxidant effects due to formation of chain initiating radicals or hormesis phenomena [89–91]. The tendency to restore cellular redox equilibrium may explain paradoxical inhibition of apoptosis by pro-oxidant interventions [92].
Location and targets of oxidative stress
There are multiple sources of reactive oxygen species in cells, representing most subcellular compartments. Mitochondria have generally been considered a principal source, although estimates range from 2–5% to 0.15% of total O2 consumption [93,94]. Superoxide generation occurs by single electron leakage to O2 at complex I, complex III and possibly the electron transfer flavoprotein-quinone oxidoreductase complex for fatty acid oxidation. Mitochondrial superoxide production is influenced by substrate fuel and respiratory state, and the site of production can be mapped by determining the effect of inhibiting electron transfer at different sites in the ETC. Superoxide generated at complex I is released at the matrix side of the inner mitochondrial membrane, and fed by forward flow of electrons from NADH or reverse flow from the complex II substrate succinate [94]. Rotenone inhibition of complex I increases superoxide produced by NADH-generating substrates, but inhibits superoxide supported by reverse electron flow [95]. Reduction of O2 at complex III is principally via the ubisemiquinone radical at center o, with release into the intermembrane space. Antimycin increases superoxide production at complex III by preventing oxidation/release of ubisemiquinone at center o, while myxothiazol inhibits superoxide by binding to center o and preventing initial oxidation of ubiquinol to ubisemiquinone.
If mitochondria-produced ROS play a critical role in apoptosis, it would be useful to know if there is a predominant site of O2− generation, either as a result of a common upstream stimulus or a required downstream target [96]. There is more support for complex I as an origin of ROS in the apoptosis literature, including TNF- or ceramide-induced and Parkinsonian models of apoptosis, with some indications that reverse electron flow may be involved [47, 97–99]. Mitochondrial complex I is inhibited by nitration and glutathionylation [100,101], as well as proteolytic inactivation by caspases and granzyme A [102,103].
Caspase-independent deaths with mitochondrial release of AIF and endonuclease G, but not cytochrome c, are usually associated with complex I-dependent superoxide production [104–108]. Whether this is related to the functional interactions between AIF, an NADH oxidase flavoprotein, and complex I (NADH dehydrogenase) activity, is unknown.
Several studies are consistent with complex III as a ROS source [109–112]. Cytochrome c, the electron acceptor reduced by complex III, also functions as a scavenger of superoxide released into the intermembrane space [113]. A significant fraction of cytochrome c, a basic protein, binds to the anionic phospholipid cardiolipin at the outer surface of the inner mitochondrial membrane, where it acquires peroxidase activity [114,115]. Cytochrome c peroxidase activity subtracts electrons from nearby unsaturated acyl chains, setting in motion progressive lipid peroxidation. Oxidation of inner mitochondrial membrane cardiolipin is an early apoptotic event required for subsequent cytochrome c release [116,117]. As lipid-soluble antioxidants can block cardiolipin oxidation, this could be a sufficient explanation for apoptosis inhibition by antioxidants. However, increased cytochrome c peroxidase activity in apoptosis is also likely to be driven by increased H2O2 substrate, which may be derived from matrix or intermembrane space superoxide due to the ability of H2O2 to cross membranes readily [96].
Other sites proposed for ROS generation during apoptosis are the plasma membrane and endoplasmic reticulum. Exposure of phosphatidylserine (PS) in the outer leaflet in the plasma membrane serves as a phagocyte recognition signal. Oxidation of PS acyl groups, as a result of the peroxidase activity of released cytochrome c, promotes externalization of PS [118]. Another ROS source, the NADPH oxidase gp91phox expressed in phagocytic cells, and homologous Nox and Duox catalytic subunits expressed in neurons, endothelial cells and other cell types, transfer cytoplasmic electrons across membranes to generate superoxide. In phagocytic cells, NADPH oxidase assembles with adaptor proteins on phagocytic vacuoles. ROS produced by NADPH oxidase have been implicated in apoptotic cell death in both phagocytic and non-phagocytic cells, usually based on inhibition of apoptosis using flavoenzyme inhibitors such as diphenylene iodonium [46, 119–122]. NADPH oxidases in non-phagocytic cells are thought to function in signaling pathways, and may also be recruited to endosomal vesicles in apoptotic cells upstream of Fas/CD95 activation [123]. There are also necrotic cell deaths linked to NADPH oxidase, and a potential role of NADPH oxidase in preventing caspase activation has been reported [124,125].
Unfolded proteins within the endoplasmic reticulum initiate stress response pathways to suppress general protein synthesis while inducing ER resident chaperones, ER membrane expansion and retrotranslocation of unfolded proteins for proteosomal degradation [126]. The unfolded protein response (UPR) also includes increased ROS generation in the ER, perhaps by an increased load on the oxidative protein folding pathway or increased GSSG/GSH ratio from reducing misplaced protein disulfides. Retrieval of oxidizable, misfolded protein substrates by ER-associated protein degradation can relieve oxidative stress within the ER [127,128]. Several ER-associated proteins with UPR functions have been shown to suppress ER-dependent ROS during apoptosis, including the Hsp70-related chaperone Grp78, TRAF2, and paraoxonase [129–131]. Endoplasmic reticulum-localized cytochrome P450 systems are also a potential source of ROS [132–136]. Mitochondrial ROS production is often found to increase with ER stress [128]. Specific ER targets of ROS relevant to apoptosis are largely unknown, although the sarco/endoplasmic reticulum Ca-ATPase (SERCA) is regulated by NO-dependent S-glutathionylation and subject to non-reversible oxidation [137].
Oxidative stress and Bcl-2
Bcl-2 was initially reported to have anti-oxidant effects in 1993 [138, 139]. Apoptotic deaths induced by dexamethasone and growth factor deprivation exhibited progressive lipid peroxidation that was completely suppressed with Bcl-2 transfection. As addition of anti-oxidants also deterred apoptosis, oxidative stress appeared causal rather than secondary to apoptosis in these experiments. Unexpectedly, no increase in ROS accounting for the increased lipid peroxidation could be detected in these models using fluorescent reporters of O2− and H2O2 or measurement of cyanide-resistant respiration. Bcl-2 also rescued cells treated with menadione, without affecting cyanide-resistant O2 consumption by bioreductive metabolism of this quinone. Similar findings were reported with t-BuOOH [140]. These results suggested that Bcl-2-expressing cells are intrinsically resistant to added oxidative stress, without directly lowering ROS levels. Conversely, apoptotic stimuli could increase oxidative damage, apparently due to endogenous (i.e. normal) production of ROS.
Cellular ROS levels can be decreased by Bcl-2. In cells depleted of glutathione, Bcl-2 expression reduced intracellular ROS levels, consistent with an effect on ROS clearance [139]. Lower ROS levels are also observed in Bcl-2-expressing cells treated with arsenic trioxide, an inhibitor of thioredoxin reductases [141]. Bcl-2 has been reported to increase cell levels or shift intracellular distribution of reduced glutathione [142–145] and a recent paper described direct binding of glutathione to Bcl-2 [146]. However, Bcl-2 does not appear to substitute for glutathione, as Bcl-2-mediated survival can be negated if reduced glutathione is sufficiently depleted in media lacking cysteine and methionine [147, 148].
Antioxidant effects of Bcl-2 should be evident in non-apoptotic contexts, for consideration as a primary or proximate function of Bcl-2. One example of this type is the observation that hyperglycemia-induced lipid peroxidation and advanced glycation end products (AGE) in endothelial cells are suppressed by Bcl-2 [149]. Unlike the classic antioxidants desferoxamine, α-tocopherol and dimethylsulfoxide, which inhibit ROS generation as well as lipid peroxidation/AGE formation, ROS levels were unaffected by Bcl-2 in these studies. Perhaps as a related phenomenon, Bcl-2 was shown to increase the activity of plasma membrane Na+/K+-ATPase, a redox-sensitive ion pump in unstimulated cells [150, 151]. The anti-oxidant phenotype with Bcl-2 expression in mammalian cells is also demonstrated by heterologous expression of Bcl-2 in Saccharomyces cerevisiae, an organism lacking Bcl-2 orthologs. Yeast transformed with expression plasmids for Bcl-2, Bcl-xL or the C. elegans ortholog CED-9 are resistant to oxidative stress, while SOD-deficient strains of the budding yeast, are partially rescued by Bcl-2 [139, 152].
Bcl-2−/− mice have increased content of oxidized proteins in multiple tissues [153,154]. Retinal endothelial cells from these mice also exhibit in vitro defects in capillary morphogenesis that can be restored by treatment with N-acetylcysteine [155]. Conversely, Bax expression in mammalian and heterologous models increases ROS generation and oxidative damage [156–162].
An opposite effect of Bcl-2 to increase cellular levels of ROS has been reported in HL-60 cells depleted of glutathione [163,164], bacteria [165], astrocytes [166], several cancer cell lines [167,168], and fibroblasts [169]. These observations have led to the proposal that Bcl-2 produces a mild increase in cellular oxidative stress, leading to increased anti-oxidant capacity within the cell and better ability to withstand additional oxidative stress. Such effects may also be related to expression level [170,171].
An interesting perspective on the relationship between Bcl-2, ROS and mitochondrial function was recently presented in relation to treatment with mitochondrial complex inhibitors [172]. CEM lymphoblastic leukemia cells and HeLa cells transfected with Bcl-2 were noted to have higher rates of respiration and cytochrome oxidase activity than control cells, along with higher mitochondrial production of O2− using the MitoSOX Red fluorescent reporter. Therefore, cellular ROS levels may normally increase with a shift toward mitochondrial ATP production. Treatment with rotenone or antimycin A (complex I and III inhibitors, respectively) increased mitochondrial O2− generation in control cells, but not Bcl-2-expressing cells. The flat O2− response in Bcl-2-expressing mitochondria was associated with depressed cytochrome oxidase activity, suggesting to the authors that Bcl-2 prevented excessive mitochondrial ROS production by reducing mitochondrial ETC activity.
As discussed above, mitochondrial effects of Bcl-2 are not restricted to limiting ROS production. Alterations in mitochondrial membrane potential (MMP) are commonly observed in apoptotic cells, most often membrane depolarization that can occur early or delayed relative to other apoptotic changes [173]. In contrast, several groups have described an early increase in MMP after apoptotic stimuli including camptothecin, staurosporine, Fas ligation, glucose and IL-3 withdrawal [174–180]. Notably, Vander Heiden et al. demonstrated that Bcl-xL prevents mitochondrial hyperpolarization in the setting of IL-3 withdrawal [175]. Apoptosis induced by growth factor deprivation is associated with decreased outer mitochondrial membrane permeability, resulting in impaired ATP/ADP exchange. In the presence of Bcl-xL, adenine nucleotide exchange is restored. Experiments with isolated mitochondria and synthetic lipid membranes indicate that Bcl-xL maintains an open conformation of the outer membrane voltage-dependent anion channel (VDAC).
Loss of outer mitochondrial membrane permeability in the presence of adequate substrates to reduce electron carriers in the respiratory chain, is expected to increase mitochondrial O2− generation. Ubisemiquinone radicals at center o in complex III are stabilized at the higher membrane potentials that are no longer used for ATP synthesis. The anti-oxidant effect of Bcl-2 might be explained as an indirect consequence of promoting adenine nucleotide exchange. However, VDAC closure is not observed with other apoptotic cues [24], including ischemia-reperfusion injury in which Bcl-2 appears to have the opposite effect of decreasing outer mitochondrial membrane permeability [181].
The availability of small molecule inhibitors for Bcl-2 and Bcl-xL affords a unique opportunity to observe the effect of acutely interrupting survival function on mitochondrial physiology [182]. By examining transfected cells overexpressing Bcl-2/Bcl-xL, responses due to inhibition of protein function can be isolated from those associated with removal of an essential survival factor. Treatment of a murine hepatocyte cell line overexpressing Bcl-xL with the Bcl-xL inhibitor, 2-methoxy antimycin A (2-MeAA), led to a progressive increase in mitochondrial membrane potential measured by TMRM fluorescence after 20–30 min. Image analysis of mitochondrial and cytosolic NAD(P)H using 2-photon excitation demonstrated a concurrent elevation in mitochondrial NAD(P)H. This response could be explained by inhibition of ATP/ADP exchange or increased substrate-driven NAD(P)H availability. In favor of the latter interpretation, fluorescence recovery after photobleaching (FRAP) studies demonstrate that the rate of NAD(P)H regeneration in mitochondria increases after 2-MeAA treatment and O2 consumption rate is unchanged at the time of initial increase in mitochondrial NAD(P)H (unpublished data).
These results suggest that Bcl-xL may “unload” the mitochondrial respiratory chain, thereby lessening O2− generation, or reductive stress within mitochondria. Bcl-xL-transfected TAMH cells are shifted toward glycolytic metabolism compared to control transfectants, with increased lactate production and reduced O2 consumption. Similar phenotypes were observed in mice expressing a Bcl-xL transgene in pancreatic islet β-cells, resulting in reduced insulin responses to hyperglycemia, tet-inducible constructs in PC-12 cells, and HeLa cells [183–185]. Notably, this phenotype is consistent with the Warburg effect, suggesting that Bcl-2 survival factors contribute to this characteristic feature of cancer cell metabolism.
By acting as a “safety valve” for mitochondrial electron transport chain flux, Bcl-2 may also improve mitochondrial function by reducing chronic oxidative stress. In some cases, this could be manifest as an increase in mitochondrial respiration, as observed in some studies. Examples of this functionality include uncoupling proteins in mammalian species and alternative oxidases in plants and lower eukaryote species [186, 187].
Bcl-2 lacks sequence or structural features of known anti-oxidant or redox active proteins, suggesting that anti-oxidant effects are indirect. The most likely explanation is that Bcl-2 modulates mitochondrial bioenergetics, i.e. availability of substrate fuels, electron flow, inner mitochondrial membrane leakiness or coupling to adenine nucleotides in a direction that diminishes known mechanisms of O2− generation within mitochondria. Although framing Bcl-2 function in this manner is less straightforward and more admitting of inconsistencies in the literature, final elucidation of Bcl-2’s connection to oxidative stress is likely to yield some novel insights for mitochondrial physiology.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Hanahan D, Weinberg RA. The hallmarks of cancer. Cell. 2000;100:57–70. doi: 10.1016/s0092-8674(00)81683-9. [DOI] [PubMed] [Google Scholar]
- 2.Manion MK, Hockenbery DM. Targeting BCL-2-related proteins in cancer therapy. Cancer Biol Ther. 2003;2:S105–S114. [PubMed] [Google Scholar]
- 3.Olejniczak ET, Van Sant C, Anderson MG, Wang G, Tahir SK, Sauter G, et al. Integrative genomic analysis of small-cell lung carcinoma reveals correlates of sensitivity to bcl-2 antagonists and uncovers novel chromosomal gains. Mol Cancer Res. 2007;5:331–339. doi: 10.1158/1541-7786.MCR-06-0367. [DOI] [PubMed] [Google Scholar]
- 4.Korsmeyer SJ. Bcl-2 initiates a new category of oncogenes: regulators of cell death. Blood. 1992;80:879–886. [PubMed] [Google Scholar]
- 5.Strasser A, Huang DC, Vaux DL. The role of the bcl-2/ced-9 gene family in cancer and general implications of defects in cell death control for tumourigenesis and resistance to chemotherapy. Biochim Biophys Acta. 1997;1333:F151–F178. doi: 10.1016/s0304-419x(97)00019-x. [DOI] [PubMed] [Google Scholar]
- 6.Fanidi A, Harrington EA, Evan GI. Cooperative interaction between c-myc and bcl-2 proto-oncogenes. Nature. 1992;359:554–556. doi: 10.1038/359554a0. [DOI] [PubMed] [Google Scholar]
- 7.Hockenbery D, Nuñez G, Milliman C, Schreiber RD, Korsmeyer SJ. Bcl-2 is an inner mitochondrial membrane protein that blocks programmed cell death. Nature. 1990 Nov 22;348(6299):334–336. doi: 10.1038/348334a0. [DOI] [PubMed] [Google Scholar]
- 8.Reed JC, Doctor KS, Godzik A. The domains of apoptosis: a genomics perspective. Sci STKE. 2004;2004:re9. doi: 10.1126/stke.2392004re9. [DOI] [PubMed] [Google Scholar]
- 9.Petros AM, Olejniczak ET, Fesik SW. Structural biology of the Bcl-2 family of proteins. Biochim Biophys Acta. 2004;1644:83–94. doi: 10.1016/j.bbamcr.2003.08.012. [DOI] [PubMed] [Google Scholar]
- 10.Hinds MG, Smits C, Fredericks-Short R, Risk JM, Bailey M, Huang DC, et al. Bim, Bad and Bmf: intrinsically unstructured BH3-only proteins that undergo a localized conformational change upon binding to prosurvival Bcl-2 targets. Cell Death Differ. 2007;14:128–136. doi: 10.1038/sj.cdd.4401934. [DOI] [PubMed] [Google Scholar]
- 11.Germain M, Shore GC. Cellular distribution of Bcl-2 family proteins. Sci STKE. 2003;173:pe10. doi: 10.1126/stke.2003.173.pe10. [DOI] [PubMed] [Google Scholar]
- 12.Nguyen M, Millar DG, Yong VW, Korsmeyer SJ, Shore GC. Targeting of Bcl-2 to the outer mitochondrial membrane by a COOH-terminal signal anchor sequence. J Biol Chem. 1993;268:25265–25268. [PubMed] [Google Scholar]
- 13.Schleiff E, Shore GC, Goping IS. Human mitochondrial import receptor, Tom20p. Use of glutathione to reveal specific interactions between Tom20-glutathione S-transferase and mitochondrial precursor proteins. FEBS Lett. 1997;404:314–318. doi: 10.1016/s0014-5793(97)00145-2. [DOI] [PubMed] [Google Scholar]
- 14.Shirane M, Nakayama KI. Inherent calcineurin inhibitor FKBP38 targets Bcl-2 to mitochondria and inhibits apoptosis. Nat Cell Biol. 2003;5:28–37. doi: 10.1038/ncb894. [DOI] [PubMed] [Google Scholar]
- 15.Kaufmann T, Schlipf S, Sanz J, Neubert K, Stein R, Borner C. Characterization of the signal that directs Bcl-xL, but not Bcl-2, to the outer mitochondrial membrane. J Cell Biol. 2003;160:53–64. doi: 10.1083/jcb.200210084. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Goping IS, Gross A, Lavoie JN, Nguyen M, Jemmerson R, Roth K, Korsmeyer SJ, Shore GC. Regulated targeting of BAX to mitochondria. J Cell Biol. 1998;143:207–215. doi: 10.1083/jcb.143.1.207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Ruffolo SC, Breckenridge DG, Nguyen M, Goping IS, Gross A, Korsmeyer SJ, et al. BID-dependent and BID-independent pathways for BAX insertion into mitochondria. Cell Death Differ. 2000;7:1101–1108. doi: 10.1038/sj.cdd.4400739. [DOI] [PubMed] [Google Scholar]
- 18.Lalier L, Cartron P-F, Juin P, Nedelkina S, Manon S, Bechinger B, et al. Bax activation and mitochondrial insertion during apoptosis. Apoptosis. 2007;12:887–896. doi: 10.1007/s10495-007-0749-1. [DOI] [PubMed] [Google Scholar]
- 19.Antonsson B, Montessuit S, Sanchez B, Martinou JC. Bax is present as a high molecular weight oligomer/complex in the mitochondrial membrane of apoptotic cells. J Biol Chem. 2001;276:11615–11623. doi: 10.1074/jbc.M010810200. [DOI] [PubMed] [Google Scholar]
- 20.Yethon JA, Epand RF, Leber B, Epand RM, Andrews DW. Interaction with a membrane surface triggers a reversible conformational change in Bax normally associated with induction of apoptosis. J Biol Chem. 2003;278:48935–48941. doi: 10.1074/jbc.M306289200. [DOI] [PubMed] [Google Scholar]
- 21.Letai A, Bassik MC, Walensky LD, Sorcinelli MD, Weiler S, Korsmeyer SJ. Distinct BH3 domains either sensitize or activate mitochondrial apoptosis, serving as prototype cancer therapeutics. Cancer Cell. 2002;2:183–192. doi: 10.1016/s1535-6108(02)00127-7. [DOI] [PubMed] [Google Scholar]
- 22.Murphy KM, Ranganathan V, Farnsworth ML, Kavallaris M, Lock RB. Bcl-2 inhibits Bax translocation from cytosol to mitochondria during drug-induced apoptosis of human tumor cells. Cell Death Differ. 2000;7:102–111. doi: 10.1038/sj.cdd.4400597. [DOI] [PubMed] [Google Scholar]
- 23.Murphy KM, Streips UN, Lock RB. Bcl-2 inhibits a Fas-induced conformational change in the Bax N terminus and Bax mitochondrial translocation. J Biol Chem. 2000;275:17225–17228. doi: 10.1074/jbc.C900590199. [DOI] [PubMed] [Google Scholar]
- 24.Vander Heiden MG, Chandel NS, Li XX, Schumacker PT, Colombinin M, Thompson CB. Outer mitochondrial membrane permeability can regulate coupled respiration and cell survival. Proc Natl Acad Sci USA. 2000;97:4666–4671. doi: 10.1073/pnas.090082297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Vander Heiden MG, Li XX, Gottlieb E, Hill RB, Thompson CB, Colombini M. Bcl-xL promotes the open configuration of the voltage-dependent anion channel and metabolite passage through the outer mitochondrial membrane. J Biol Chem. 2001;276:19414–19419. doi: 10.1074/jbc.M101590200. [DOI] [PubMed] [Google Scholar]
- 26.Zamzani N, Susin SA, Marchetti P, Hirsch T, Gomez-Monterrey I, Castedo M, et al. Mitochondrial control of nuclear apoptosis. J Exp Med. 1996;183:1533–1544. doi: 10.1084/jem.183.4.1533. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Baines CP, Kaiser RA, Purcell NH, Blair NS, Osinska H, Hambleton MA, et al. Loss of cyclophilin D reveals a critical role for mitochondrial permeability transition in cell death. Nature. 2005;434:658–662. doi: 10.1038/nature03434. [DOI] [PubMed] [Google Scholar]
- 28.Nakagawa T, Shimizu S, Watanabe T, Yamaguchi O, Otsu K, Yamagata H, et al. Cyclophilin D-dependent mitochondrial permeability transition regulates some necrotic but not apoptotic cell death. Nature. 2005;434:652–658. doi: 10.1038/nature03317. [DOI] [PubMed] [Google Scholar]
- 29.Chen R, Valencia I, Zhong F, McColl KS, Roderick HL, Bootman MD, et al. Bcl-2 functionally interacts with inositol 1,4,5-trisphosphate receptors to regulate calcium release from the ER in response to inositol 1,4,5-trisphosphate. J Cell Biol. 2004;166:193–203. doi: 10.1083/jcb.200309146. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Dremina ES, Sharov VS, Kumar K, Zaidi A, Michaelis EK, Schoneich C. Anti-apoptotic protein Bcl-2 interacts with and destabilizes the sarcoplasmic/endoplasmic reticulum Ca2+ATPase (SERCA) Biochem J. 2004;383:361–370. doi: 10.1042/BJ20040187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Oakes SA, Scorrano L, Opferman JT, Bassik MC, Nishino M, Pozzan T, et al. Proapoptotic BAX and BAK regulate the type 1 inositol trisphosphate receptor and calcium leak from the endoplasmic reticulum. Proc Natl Acad Sci USA. 2005;102:105–110. doi: 10.1073/pnas.0408352102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Hockenbery DM, Oltvai ZN, Yin XM, Milliman CL, Korsmeyer SJ. Bcl-2 functions in an antioxidant pathway to prevent apoptosis. Cell. 1993;75:241–251. doi: 10.1016/0092-8674(93)80066-n. [DOI] [PubMed] [Google Scholar]
- 33.Wolfe JT, Ross D, Cohen GM. A role for metals and free radicals in the induction of apoptosis in thymocytes. FEBS Lett. 1994;352:58–62. doi: 10.1016/0014-5793(94)00920-1. [DOI] [PubMed] [Google Scholar]
- 34.Torres-Roca JF, Lecoeur H, Amatore C, Gougeon ML. The early intracellular production of a reactive oxygen intermediate mediates apoptosis in dexamethasone-treated thymocytes. Cell Death Differ. 1995;2:309–319. [PubMed] [Google Scholar]
- 35.Tome ME, Baker AF, Powis G, Payne CM, Briehl MM. Catalase-overexpressing thymocytes are resistant to glucocorticoid-induced apoptosis and exhibit increased net tumor growth. Cancer Res. 2001;61:2766–2773. [PubMed] [Google Scholar]
- 36.Abello PA, Fidler SA, Buchman TG. Thiol reducing agents modulate induced apoptosis in porcine endothelial cells. Shock. 1994;2:79–83. doi: 10.1097/00024382-199408000-00001. [DOI] [PubMed] [Google Scholar]
- 37.Rauen U, Petrat F, Li T, De Groot H. Hypothermia injury/cold-induced apoptosis--evidence of an increase in chelatable iron causing oxidative injury in spite of low O2-/H2O2 formation. FASEB J. 2000;14:1953–1964. doi: 10.1096/fj.00-0071com. [DOI] [PubMed] [Google Scholar]
- 38.Shimura M, Osawa Y, Yuo A, Hatake K, Takaku F, Ishizaka Y. Oxidative stress as a necessary factor in room temperature-induced apoptosis of HL-60 cells. J Leukoc Biol. 2000;68:87–96. [PubMed] [Google Scholar]
- 39.Khar A, Pardhasaradhi BV, Ali AM, Kumari AL. Protection conferred by Bcl-2 expression involves reduced oxidative stress and increased glutathione production during hypothermia-induced apoptosis in AK-5 tumor cells. Free Radic Biol Med. 2003;35:949–957. doi: 10.1016/s0891-5849(03)00469-6. [DOI] [PubMed] [Google Scholar]
- 40.Shin SW, Kil IS, Park JW. Silencing of mitochondrial NADP+dependent isocitrate dehydrogenase by small interfering RNA enhances heat shock-induced apoptosis. Biochem Biophys Res Commun. 2008;366:1012–1018. doi: 10.1016/j.bbrc.2007.12.067. [DOI] [PubMed] [Google Scholar]
- 41.Greenlund LJ, Deckwerth TL, Johnson EM., Jr Superoxide dismutase delays neuronal apoptosis: a role for reactive oxygen species in programmed neuronal death. Neuron. 1995;14:303–315. doi: 10.1016/0896-6273(95)90287-2. [DOI] [PubMed] [Google Scholar]
- 42.Ratan RR, Lee PJ, Baraban JM. Serum deprivation inhibits glutathione depletion-induced death in embryonic cortical neurons: evidence against oxidative stress as a final common mediator of neuronal apoptosis. Neurochem Int. 1996;29:153–157. doi: 10.1016/0197-0186(95)00115-8. [DOI] [PubMed] [Google Scholar]
- 43.Barroso MP, Gómez-Diaz C, Lopez-Lluch G, Malagón MM, Crane FL, Navas P. Ascorbate and alpha-tocopherol prevent apoptosis induced by serum removal independent of Bcl-2. Arch Biochem Biophys. 1997;343:243–248. doi: 10.1006/abbi.1997.0170. [DOI] [PubMed] [Google Scholar]
- 44.Lieberthal W, Triaca V, Koh JS, Pagano PJ, Levine JS. Role of superoxide in apoptosis induced by growth factor withdrawal. Am J Physiol. 1998;275:F691–F702. doi: 10.1152/ajprenal.1998.275.5.F691. [DOI] [PubMed] [Google Scholar]
- 45.López-Lluch G, Barroso MP, Martín SF, Fernández-Ayala DJ, Gómez-Díaz C, Villalba JM, et al. Role of plasma membrane coenzyme Q on the regulation of apoptosis. Biofactors. 1999;9:171–177. doi: 10.1002/biof.5520090212. [DOI] [PubMed] [Google Scholar]
- 46.Tammariello SP, Quinn MT, Estus S. NADPH oxidase contributes directly to oxidative stress and apoptosis in nerve growth factor-deprived sympathetic neurons. J Neurosci. 2000;20:RC53. doi: 10.1523/JNEUROSCI.20-01-j0006.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Schulze-Osthoff K, Bakker AC, Vanhaesebroeck B, Beyaert R, Jacob WA, Fiers W. Cytotoxic activity of tumor necrosis factor is mediated by early damage of mitochondrial functions. Evidence for the involvement of mitochondrial radical generation. J Biol Chem. 1992;267:5317–5323. [PubMed] [Google Scholar]
- 48.O’Donnell VB, Spycher S, Azzi A. Involvement of oxidants and oxidant-generating enzyme(s) in tumour-necrosis-factor-alpha-mediated apoptosis: role for lipoxygenase pathway but not mitochondrial respiratory chain. Biochem J. 1995;310:133–141. doi: 10.1042/bj3100133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Um HD, Orenstein JM, Wahl SM. Fas mediates apoptosis in human monocytes by a reactive oxygen intermediate dependent pathway. J Immunol. 1996;156:3469–3477. [PubMed] [Google Scholar]
- 50.Kasahara Y, Iwai K, Yachie A, Ohta K, Konno A, Seki H, et al. Involvement of reactive oxygen intermediates in spontaneous and CD95 (Fas/APO-1)-mediated apoptosis of neutrophils. Blood. 1997;89(5):1748–1753. [PubMed] [Google Scholar]
- 51.Toborek M, Blanc EM, Kaiser S, Mattson MP, Hennig B. Linoleic acid potentiates TNF-mediated oxidative stress, disruption of calcium homeostasis, and apoptosis of cultured vascular endothelial cells. J Lipid Res. 1997;38:2155–2167. [PubMed] [Google Scholar]
- 52.Petrache I, Otterbein LE, Alam J, Wiegand GW, Choi AM. Heme oxygenase-1 inhibits TNF-alpha-induced apoptosis in cultured fibroblasts. Am J Physiol Lung Cell Mol Physiol. 2000;278:L312–L319. doi: 10.1152/ajplung.2000.278.2.L312. [DOI] [PubMed] [Google Scholar]
- 53.Pierce RH, Campbell JS, Stephenson AB, Franklin CC, Chaisson M, Poot M, et al. Disruption of redox homeostasis in tumor necrosis factor-induced apoptosis in a murine hepatocyte cell line. Am J Pathol. 2000;157:221–236. doi: 10.1016/S0002-9440(10)64533-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Gottlieb E, Vander Heiden MG, Thompson CB. Bcl-x(L) prevents the initial decrease in mitochondrial membrane potential and subsequent reactive oxygen species production during tumor necrosis factor alpha-induced apoptosis. Mol Cell Biol. 2000;20:5680–5689. doi: 10.1128/mcb.20.15.5680-5689.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Chen J, Li D, Zhang X, Mehta JL. Tumor necrosis factor-alpha-induced apoptosis of human coronary artery endothelial cells: modulation by the peroxisome proliferator-activated receptor-gamma ligand pioglitazone. J Cardiovasc Pharmacol Ther. 2000;9:35–41. doi: 10.1177/107424840400900i106. [DOI] [PubMed] [Google Scholar]
- 56.Kagan VE, Gleiss B, Tyurina YY, Tyurin VA, Elenström-Magnusson C, Liu SX, et al. A role for oxidative stress in apoptosis: oxidation and externalization of phosphatidylserine is required for macrophage clearance of cells undergoing Fas-mediated apoptosis. J Immunol. 2002;169:487–499. doi: 10.4049/jimmunol.169.1.487. [DOI] [PubMed] [Google Scholar]
- 57.Lee MW, Park SC, Kim JH, Kim IK, Han KS, Kim KY, et al. The involvement of oxidative stress in tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL)-induced apoptosis in HeLa cells. Cancer Lett. 2002;182:75–82. doi: 10.1016/s0304-3835(02)00074-5. [DOI] [PubMed] [Google Scholar]
- 58.Gouaze V, Andrieu-Abadie N, Cuvillier O, Malagarie-Cazenave S, Frisach MF, Mirault ME, et al. Glutathione peroxidase-1 protects from CD95-induced apoptosis. J Biol Chem. 2002;277:42867–42874. doi: 10.1074/jbc.M203067200. [DOI] [PubMed] [Google Scholar]
- 59.Devadas S, Hinshaw JA, Zaritskaya L, Williams MS. Fas-stimulated generation of reactive oxygen species or exogenous oxidative stress sensitize cells to Fas-mediated apoptosis. Free Radic Biol Med. 2003;35:648–661. doi: 10.1016/s0891-5849(03)00391-5. [DOI] [PubMed] [Google Scholar]
- 60.Perez-Cruz I, Cárcamo JM, Golde DW. Caspase-8 dependent TRAIL-induced apoptosis in cancer cell lines is inhibited by vitamin C and catalase. Apoptosis. 2007;12:225–234. doi: 10.1007/s10495-006-0475-0. [DOI] [PubMed] [Google Scholar]
- 61.Jin S, Ray RM, Johnson LR. TNF-alpha/cycloheximide-induced apoptosis in intestinal epithelial cells requires Rac1-regulated reactive oxygen species. Am J Physiol Gastrointest Liver Physiol. 2008;294:G928–G937. doi: 10.1152/ajpgi.00219.2007. [DOI] [PubMed] [Google Scholar]
- 62.Malorni W, Rivabene R, Santini MT, Donelli G. N-acetylcysteine inhibits apoptosis and decreases viral particles in HIV-chronically infected U937 cells. FEBS Lett. 1993;327:75–78. doi: 10.1016/0014-5793(93)81043-y. [DOI] [PubMed] [Google Scholar]
- 63.Lin KI, Lee SH, Narayanan R, Baraban JM, Hardwick JM, Ratan RR. Thiol agents and Bcl-2 identify an alphavirus-induced apoptotic pathway that requires activation of the transcription factor NF-kappa B. J Cell Biol. 1995;131:1149–1161. doi: 10.1083/jcb.131.5.1149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Xu Y, Nguyen Q, Lo DC, Czaja MJ. c-myc-Dependent hepatoma cell apoptosis results from oxidative stress and not a deficiency of growth factors. J Cell Physiol. 1997;170:192–199. doi: 10.1002/(SICI)1097-4652(199702)170:2<192::AID-JCP11>3.0.CO;2-K. [DOI] [PubMed] [Google Scholar]
- 65.Lotem J, Peled-Kamar M, Groner Y, Sachs L. Cellular oxidative stress and the control of apoptosis by wild-type p53, cytotoxic compounds, and cytokines. Proc Natl Acad Sci U S A. 1996;93:9166–9171. doi: 10.1073/pnas.93.17.9166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Polyak K, Xia Y, Zweier JL, Kinzler KW, Vogelstein B. A model for p53-induced apoptosis. Nature. 1997 Sep;389:300–305. doi: 10.1038/38525. [DOI] [PubMed] [Google Scholar]
- 67.Drane P, Bravard A, Bouvard V, May E. Reciprocal down-regulation of p53 and SOD2 gene expression-implication in p53 mediated apoptosis. Oncogene. 2001;20:430–439. doi: 10.1038/sj.onc.1204101. [DOI] [PubMed] [Google Scholar]
- 68.Hwang PM, Bunz F, Yu J, Rago C, Chan TA, Murphy MP, et al. Ferredoxin reductase affects p53-dependent, 5-fluorouracil-induced apoptosis in colorectal cancer cells. Nat Med. 2001;7:1111–1117. doi: 10.1038/nm1001-1111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Kruman I, Guo Q, Mattson MP. Calcium and reactive oxygen species mediate staurosporine-induced mitochondrial dysfunction and apoptosis in PC12 cells. J Neurosci Res. 1998;51:293–308. doi: 10.1002/(SICI)1097-4547(19980201)51:3<293::AID-JNR3>3.0.CO;2-B. [DOI] [PubMed] [Google Scholar]
- 70.Patel M. Inhibition of neuronal apoptosis by a metalloporphyrin superoxide dismutase mimic. J Neurochem. 1998;71:1068–1074. doi: 10.1046/j.1471-4159.1998.71031068.x. [DOI] [PubMed] [Google Scholar]
- 71.Nomura K, Imai H, Koumura T, Arai M, Nakagawa Y. Mitochondrial phospholipid hydroperoxide glutathione peroxidase suppresses apoptosis mediated by a mitochondrial death pathway. J Biol Chem. 1999;274:29294–29302. doi: 10.1074/jbc.274.41.29294. [DOI] [PubMed] [Google Scholar]
- 72.Hoye AT, Davoren JE, Wipf P, Fink MP, Kagan VE. Targeting mitochondria. Acc Chem Res. 2008;41:87–97. doi: 10.1021/ar700135m. [DOI] [PubMed] [Google Scholar]
- 73.Jacobson MD, Raff MC. Programmed cell death and Bcl-2 protection in very low oxygen. Nature. 1995;374:814–816. doi: 10.1038/374814a0. [DOI] [PubMed] [Google Scholar]
- 74.Shimizu S, Eguchi Y, Kosaka H, Kamiike W, Matsuda H, Tsujimoto Y. Prevention of hypoxia-induced cell death by Bcl-2 and Bcl-xL. Nature. 1995;374:811–813. doi: 10.1038/374811a0. [DOI] [PubMed] [Google Scholar]
- 75.Degli Esposti M, McLennan H. Mitochondria and cells produce reactive oxygen species in virtual anaerobiosis: relevance to ceramide-induced apoptosis. FEBS Lett. 1998;430:338–342. doi: 10.1016/s0014-5793(98)00688-7. [DOI] [PubMed] [Google Scholar]
- 76.Guzy RD, Schumacker PT. Oxygen sensing by mitochondria at complex III: the paradox of increased reactive oxygen species during hypoxia. Exp Physiol. 2006;91:807–819. doi: 10.1113/expphysiol.2006.033506. [DOI] [PubMed] [Google Scholar]
- 77.Rumsey WL, Schlosser C, Nuutinen EM, Robiolio M, Wilson DF. Cellular energetics and the oxygen dependence of respiration in cardiac myocytes isolated from adult rat. J Biol Chem. 1990;265:15392–15399. [PubMed] [Google Scholar]
- 78.Wilson DF, Rumsey WL, Green TJ, Vanderkooi JM. The oxygen dependence of mitochondrial oxidative phosphorylation measured by a new optical method for measuring oxygen concentration. J Biol Chem. 1988;263:2712–2718. [PubMed] [Google Scholar]
- 79.Chandel NS, Budinger GRS, Schumacker PT. Molecular oxygen modulates cytochrome c oxidase function. J Biol Chem. 1996;271:18672–18677. doi: 10.1074/jbc.271.31.18672. [DOI] [PubMed] [Google Scholar]
- 80.Guzy RD, Hoyos B, Robin E, Chen H, Liu L, Mansfield KD, et al. Mitochondrial complex III is required for hypoxia-induced ROS production and cellular oxygen sensing. Cell Metab. 2005;1:401–408. doi: 10.1016/j.cmet.2005.05.001. [DOI] [PubMed] [Google Scholar]
- 81.Brunelle JK, Bell EL, Quesada NM, Vercauteren K, Tiranti V, Zeviani M, et al. Oxygen sensing requires mitochondrial ROS but not oxidative phosphorylation. Cell Metab. 2005;1:409–414. doi: 10.1016/j.cmet.2005.05.002. [DOI] [PubMed] [Google Scholar]
- 82.McClintock DS, Santore MT, Lee VY, Brunelle J, Budinger GR, Zong WX, et al. Bcl-2 family members and functional electron transport chain regulate oxygen deprivation-induced cell death. Mol Cell Biol. 2002;22:94–104. doi: 10.1128/MCB.22.1.94-104.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Allen CB, Schneider BK, White CW. Limitations to oxygen diffusion and equilibration in in vitro cell exposure systems in hyperoxia and hypoxia. Am J Physiol Lung Cell Mol Physiol. 2001;281:L1021–L1027. doi: 10.1152/ajplung.2001.281.4.L1021. [DOI] [PubMed] [Google Scholar]
- 84.Koch CJ, Kruuv J. The release of oxygen from polystyrene Petri dishes. Br J Radiol. 1972 Oct;45(538):787–788. doi: 10.1259/0007-1285-45-538-787. [DOI] [PubMed] [Google Scholar]
- 85.Kollau A, Beretta M, Russwurm M, Koesling D, Keung WM, Schmidt K, et al. Mitochondrial nitrite reduction coupled to soluble guanylate cyclase activation: lack of evidence for a role in the bioactivation of nitroglycerin. Nitric Oxide. 2009;20:53–60. doi: 10.1016/j.niox.2008.09.003. [DOI] [PubMed] [Google Scholar]
- 86.Igamberdiev AU, Hill RD. Plant Mitochondrial Function During Anaerobiosis. Ann Bot (Lond) 2008 doi: 10.1093/aob/mcn100. [Epub ahead of print] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Hug H, Enari M, Nagata S. No requirement of reactive oxygen intermediates in Fas-mediated apoptosis. FEBS Lett. 1994;351:311–313. doi: 10.1016/0014-5793(94)00852-3. [DOI] [PubMed] [Google Scholar]
- 88.Sentürker S, Tschirret-Guth R, Morrow J, Levine R, Shacter E. Induction of apoptosis by chemotherapeutic drugs without generation of reactive oxygen species. Arch Biochem Biophys. 2002;397:262–272. doi: 10.1006/abbi.2001.2681. [DOI] [PubMed] [Google Scholar]
- 89.Sagristá ML, García AE, Africa De Madariaga M, Mora M. Antioxidant and pro-oxidant effect of the thiolic compounds N-acetyl-L-cysteine and glutathione against free radical-induced lipid peroxidation. Free Radic Res. 2002;36:329–340. doi: 10.1080/10715760290019354. [DOI] [PubMed] [Google Scholar]
- 90.Budinger GR, Tso M, McClintock DS, Dean DA, Sznajder JI, Chandel NS. Hyperoxia-induced apoptosis does not require mitochondrial reactive oxygen species and is regulated by Bcl-2 proteins. J Biol Chem. 2002;277:15654–15660. doi: 10.1074/jbc.M109317200. [DOI] [PubMed] [Google Scholar]
- 91.McCord JM. Superoxide dismutase, lipid peroxidation, and bell-shaped dose response curves. Dose Response. 2008;6:223–238. doi: 10.2203/dose-response.08-012.McCord. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Clément MV, Ponton A, Pervaiz S. Apoptosis induced by hydrogen peroxide is mediated by decreased superoxide anion concentration and reduction of intracellular milieu. FEBS Lett. 1998;440:13–18. doi: 10.1016/s0014-5793(98)01410-0. [DOI] [PubMed] [Google Scholar]
- 93.Halliwell B, Gutteridge JMC. Free Radical Biology and Medicine. Oxford, UK: Oxford University Press; 1989. [Google Scholar]
- 94.St-Pierre J, Buckingham JA, Roebuck SJ, Brand MD. Topology of superoxide production from different sites in the mitochondrial electron transport chain. J Biol Chem. 2002;277:44784–44790. doi: 10.1074/jbc.M207217200. [DOI] [PubMed] [Google Scholar]
- 95.Liu Y, Fiskum G, Schubert D. Generation of reactive oxygen species by the mitochondrial electron transport chain. J Neurochem. 2002;80:780–787. doi: 10.1046/j.0022-3042.2002.00744.x. [DOI] [PubMed] [Google Scholar]
- 96.Jiang J, Huang Z, Zhao Q, Feng W, Belikova NA, Kagan VE. Interplay between bax, reactive oxygen species production, and cardiolipin oxidation during apoptosis. Biochem Biophys Res Commun. 2008;368:145–150. doi: 10.1016/j.bbrc.2008.01.055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.France-Lanord V, Brugg B, Michel PP, Agid Y, Ruberg M. Mitochondrial free radical signal in ceramide-dependent apoptosis: a putative mechanism for neuronal death in Parkinson's disease. J Neurochem. 1997;69:1612–1621. doi: 10.1046/j.1471-4159.1997.69041612.x. [DOI] [PubMed] [Google Scholar]
- 98.Quillet-Mary A, Jaffrézou JP, Mansat V, Bordier C, Naval J, Laurent G. Implication of mitochondrial hydrogen peroxide generation in ceramide-induced apoptosis. J Biol Chem. 1997;272:21388–21395. doi: 10.1074/jbc.272.34.21388. [DOI] [PubMed] [Google Scholar]
- 99.Goossens V, Stangé G, Moens K, Pipeleers D, Grooten J. Regulation of tumor necrosis factor-induced, mitochondria- and reactive oxygen species-dependent cell death by the electron flux through the electron transport chain complex I. Antioxid Redox Signal. 1999;1:285–295. doi: 10.1089/ars.1999.1.3-285. [DOI] [PubMed] [Google Scholar]
- 100.Yamamoto T, Maruyama W, Kato Y, Yi H, Shamoto-Nagai M, Tanaka M, et al. Selective nitration of mitochondrial complex I by peroxynitrite: involvement in mitochondria dysfunction and cell death of dopaminergic SH-SY5Y cells. J Neural Transm. 2002;109:1–13. doi: 10.1007/s702-002-8232-1. [DOI] [PubMed] [Google Scholar]
- 101.Taylor ER, Hurrell F, Shannon RJ, Lin TK, Hirst J, Murphy MP. Reversible glutathionylation of complex I increases mitochondrial superoxide formation. J Biol Chem. 2003;278:19603–19610. doi: 10.1074/jbc.M209359200. [DOI] [PubMed] [Google Scholar]
- 102.Ricci JE, Muñoz-Pinedo C, Fitzgerald P, Bailly-Maitre B, Perkins GA, Yadava N, et al. Disruption of mitochondrial function during apoptosis is mediated by caspase cleavage of the p75 subunit of complex I of the electron transport chain. Cell. 2004;117:773–786. doi: 10.1016/j.cell.2004.05.008. [DOI] [PubMed] [Google Scholar]
- 103.Martinvalet D, Dykxhoorn DM, Ferrini R, Lieberman J. Granzyme A cleaves a mitochondrial complex I protein to initiate caspase-independent cell death. Cell. 2008;133:681–692. doi: 10.1016/j.cell.2008.03.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Dumont C, Dürrbach A, Bidère N, Rouleau M, Kroemer G, Bernard G, et al. Caspase-independent commitment phase to apoptosis in activated blood T lymphocytes: reversibility at low apoptotic insult. Blood. 2000;96:1030–1038. [PubMed] [Google Scholar]
- 105.Candé C, Cohen I, Daugas E, Ravagnan L, Larochette N, Zamzami N, et al. Apoptosis-inducing factor (AIF): a novel caspase-independent death effector released from mitochondria. Biochimie. 2002;84:215–222. doi: 10.1016/s0300-9084(02)01374-3. [DOI] [PubMed] [Google Scholar]
- 106.Shih CM, Ko WC, Wu JS, Wei YH, Wang LF, Chang EE, et al. Mediating of caspase-independent apoptosis by cadmium through the mitochondria-ROS pathway in MRC-5 fibroblasts. J Cell Biochem. 2004;91:384–397. doi: 10.1002/jcb.10761. [DOI] [PubMed] [Google Scholar]
- 107.Santamaría G, Martínez-Diez M, Fabregat I, Cuezva JM. Efficient execution of cell death in non-glycolytic cells requires the generation of ROS controlled by the activity of mitochondrial H+-ATP synthase. Carcinogenesis. 2006;27:925–935. doi: 10.1093/carcin/bgi315. [DOI] [PubMed] [Google Scholar]
- 108.Marella M, Seo BB, Matsuno-Yagi A, Yagi T. Mechanism of cell death caused by complex I defects in a rat dopaminergic cell line. J Biol Chem. 2007;282:24146–24156. doi: 10.1074/jbc.M701819200. [DOI] [PubMed] [Google Scholar]
- 109.Gudz TI, Tserng KY, Hoppel CL. Direct inhibition of mitochondrial respiratory chain complex III by cell-permeable ceramide. J Biol Chem. 1997;272:24154–24158. doi: 10.1074/jbc.272.39.24154. [DOI] [PubMed] [Google Scholar]
- 110.Suzuki S, Higuchi M, Proske RJ, Oridate N, Hong WK, Lotan R. Implication of mitochondria-derived reactive oxygen species, cytochrome C and caspase-3 in N-(4-hydroxyphenyl)retinamide-induced apoptosis in cervical carcinoma cells. Oncogene. 1999;18:6380–6387. doi: 10.1038/sj.onc.1203024. [DOI] [PubMed] [Google Scholar]
- 111.Pham NA, Hedley DW. Respiratory chain-generated oxidative stress following treatment of leukemic blasts with DNA-damaging agents. Exp Cell Res. 2001;264:345–352. doi: 10.1006/excr.2000.5148. [DOI] [PubMed] [Google Scholar]
- 112.Nishimura G, Proske RJ, Doyama H, Higuchi M. Regulation of apoptosis by respiration: cytochrome c release by respiratory substrates. FEBS Lett. 2001;505:399–404. doi: 10.1016/s0014-5793(01)02859-9. [DOI] [PubMed] [Google Scholar]
- 113.Pereverzev MO, Vygodina TV, Konstantinov AA, Skulachev VP. Cytochrome c, an ideal antioxidant. Biochem Soc Trans. 2003;31:1312–1315. doi: 10.1042/bst0311312. [DOI] [PubMed] [Google Scholar]
- 114.Kagan VE, Borisenko GG, Tyurina YY, Tyurin VA, Jiang J, Potapovich AI, et al. Oxidative lipidomics of apoptosis: redox catalytic interactions of cytochrome c with cardiolipin and phosphatidylserine. Free Radic Biol Med. 2004;37:1963–1985. doi: 10.1016/j.freeradbiomed.2004.08.016. [DOI] [PubMed] [Google Scholar]
- 115.Iverson SL, Orrenius O. The cardiolipin-cytochrome c interaction and the mitochondrial regulation of apoptosis. Arch Biochem Biophys. 2004;423:37–46. doi: 10.1016/j.abb.2003.12.002. [DOI] [PubMed] [Google Scholar]
- 116.Kagan VE, Tyurin VA, Jiang J, Tyurina YY, Ritov VB, Amoscato AA, et al. Cytochrome c acts as a cardiolipin oxygenase required for release of proapoptotic factors. Nat Chem Biol. 2005;1:223–232. doi: 10.1038/nchembio727. [DOI] [PubMed] [Google Scholar]
- 117.Gonzalvez F, Gottlieb E. Cardiolipin: setting the beat of apoptosis. Apoptosis. 2007;12:877–885. doi: 10.1007/s10495-007-0718-8. [DOI] [PubMed] [Google Scholar]
- 118.Bayir H, Fadeel B, Palladino MJ, Witasp E, Kurnikov IV, Tyurina YY, et al. Apoptotic interactions of cytochrome c: redox flirting with anionic phospholipids within and outside of mitochondria. Biochim Biophys Acta. 2006;1757:648–659. doi: 10.1016/j.bbabio.2006.03.002. [DOI] [PubMed] [Google Scholar]
- 119.Suzuki Y, Ono Y, Hirabayashi Y. Rapid and specific reactive oxygen species generation via NADPH oxidase activation during Fas-mediated apoptosis. FEBS Lett. 1998;425:209–212. doi: 10.1016/s0014-5793(98)00228-2. [DOI] [PubMed] [Google Scholar]
- 120.Arroyo A, Modrianský M, Serinkan FB, Bello RI, Matsura T, Jiang J, et al. NADPH oxidase-dependent oxidation and externalization of phosphatidylserine during apoptosis in Me2SO-differentiated HL-60 cells Role in phagocytic clearance. J Biol Chem. 2002;277:49965–49975. doi: 10.1074/jbc.M204513200. [DOI] [PubMed] [Google Scholar]
- 121.Carmona-Cuenca I, Herrera B, Ventura JJ, Roncero C, Fernández M, Fabregat I. EGF blocks NADPH oxidase activation by TGF-beta in fetal rat hepatocytes, impairing oxidative stress, and cell death. J Cell Physiol. 2006;207:322–330. doi: 10.1002/jcp.20568. [DOI] [PubMed] [Google Scholar]
- 122.Qin F, Patel R, Yan C, Liu W. NADPH oxidase is involved in angiotensin II-induced apoptosis in H9C2 cardiac muscle cells: effects of apocynin. Free Radic Biol Med. 2006;40:236–246. doi: 10.1016/j.freeradbiomed.2005.08.010. [DOI] [PubMed] [Google Scholar]
- 123.Reinehr R, Becker S, Braun J, Eberle A, Grether-Beck S, Haüssinger D. Endosomal acidification and activation of NADPH oxidase isoforms are upstream events in hyperosmolarity-induced hepatocyte apoptosis. J Biol Chem. 2006;281:23150–23166. doi: 10.1074/jbc.M601451200. [DOI] [PubMed] [Google Scholar]
- 124.Wilkie RP, Vissers MC, Dragunow M, Hampton MB. A functional NADPH oxidase prevents caspase involvement in the clearance of phagocytic neutrophils. Infect Immun. 2007;75:3256–3263. doi: 10.1128/IAI.01984-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Kim YS, Morgan MJ, Choksi S, Liu ZG. TNF-induced activation of the Nox1 NADPH oxidase and its role in the induction of necrotic cell death. Mol Cell. 2007;26:675–687. doi: 10.1016/j.molcel.2007.04.021. [DOI] [PubMed] [Google Scholar]
- 126.Malhotra JD, Kaufman RJ. The endoplasmic reticulum and the unfolded protein response. Semin Cell Dev Biol. 2007;18:716–731. doi: 10.1016/j.semcdb.2007.09.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Harding HP, Zhang Y, Zeng H, Novoa I, Lu PD, Calfon M, et al. An integrated stress response regulates amino acid metabolism and resistance to oxidative stress. Mol Cell. 2003;11:619–633. doi: 10.1016/s1097-2765(03)00105-9. [DOI] [PubMed] [Google Scholar]
- 128.Haynes CM, Titus EA, Cooper AA. Degradation of misfolded proteins prevents ER-derived oxidative stress and cell death. Mol Cell. 2004;15:767–776. doi: 10.1016/j.molcel.2004.08.025. [DOI] [PubMed] [Google Scholar]
- 129.Yu Z, Luo H, Fu W, Mattson MP. The endoplasmic reticulum stress-responsive protein GRP78 protects neurons against excitotoxicity and apoptosis: suppression of oxidative stress and stabilization of calcium homeostasis. Exp Neurol. 1999;155:302–314. doi: 10.1006/exnr.1998.7002. [DOI] [PubMed] [Google Scholar]
- 130.Mauro C, Crescenzi E, De Mattia R, Pacifico F, Mellone S, Salzano S, et al. Central role of the scaffold protein tumor necrosis factor receptor-associated factor 2 in regulating endoplasmic reticulum stress-induced apoptosis. J Biol Chem. 2006;281:2631–2638. doi: 10.1074/jbc.M502181200. [DOI] [PubMed] [Google Scholar]
- 131.Horke S, Witte I, Wilgenbus P, Altenhöfer S, Krueger M, Li H, et al. Protective effect of paraoxonase-2 against ER stress-induced apoptosis is lost upon disturbance of calcium-homeostasis. Biochem J. 2008 doi: 10.1042/BJ20080775. [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
- 132.Chen Q, Cederbaum AI. Cytotoxicity and apoptosis produced by cytochrome P450 2E1 in Hep G2 cells. Mol Pharmacol. 1998;53:638–648. doi: 10.1124/mol.53.4.638. [DOI] [PubMed] [Google Scholar]
- 133.Shiba D, Shimamoto N. Attenuation of endogenous oxidative stress-induced cell death by cytochrome P450 inhibitors in primary cultures of rat hepatocytes. Free Radic Biol Med. 1999;27:1019–1026. doi: 10.1016/s0891-5849(99)00150-1. [DOI] [PubMed] [Google Scholar]
- 134.Haouzi D, Lekéhal M, Moreau A, Moulis C, Feldmann G, Robin MA, et al. Cytochrome P450-generated reactive metabolites cause mitochondrial permeability transition, caspase activation, and apoptosis in rat hepatocytes. Hepatology. 2000;32:303–311. doi: 10.1053/jhep.2000.9034. [DOI] [PubMed] [Google Scholar]
- 135.Gottlieb RA. Cytochrome P450: major player in reperfusion injury. Arch Biochem Biophys. 2003;420:262–267. doi: 10.1016/j.abb.2003.07.004. [DOI] [PubMed] [Google Scholar]
- 136.Liu H, Baliga R. Endoplasmic reticulum stress-associated caspase 12 mediates cisplatin-induced LLC-PK1 cell apoptosis. J Am Soc Nephrol. 2005;16:1985–1992. doi: 10.1681/ASN.2004090768. [DOI] [PubMed] [Google Scholar]
- 137.Cohen RA, Adachi T. Nitric-oxide-induced vasodilatation: regulation by physiologic s-glutathiolation and pathologic oxidation of the sarcoplasmic endoplasmic reticulum calcium ATPase. Trends Cardiovasc Med. 2006;16:109–114. doi: 10.1016/j.tcm.2006.02.001. [DOI] [PubMed] [Google Scholar]
- 138.Hockenbery DM, Oltvai ZN, Yin XM, Milliman CL, Korsmeyer SJ. Bcl-2 functions in an antioxidant pathway to prevent apoptosis. Cell. 1993;75:241–251. doi: 10.1016/0092-8674(93)80066-n. [DOI] [PubMed] [Google Scholar]
- 139.Kane DJ, Sarafian TA, Anton R, Hahn H, Gralla EB, Valentine JS, et al. Bcl-2 inhibition of neural death: decreased generation of reactive oxygen species. Science. 1993;262:1274–1277. doi: 10.1126/science.8235659. [DOI] [PubMed] [Google Scholar]
- 140.Saitoh Y, Ouchida R, Kayasuga A, Miwa N. Anti-apoptotic defense of bcl-2 gene against hydroperoxide-induced cytotoxicity together with suppressed lipid peroxidation, enhanced ascorbate uptake, and upregulated Bcl-2 protein. J Cell Biochem. 2003;89:321–334. doi: 10.1002/jcb.10506. [DOI] [PubMed] [Google Scholar]
- 141.Gupta S, Yel L, Kim D, Kim C, Chiplunkar S, Gollapudi S. Arsenic trioxide induces apoptosis in peripheral blood T lymphocyte subsets by inducing oxidative stress: a role of Bcl-2. Mol Cancer Ther. 2003;2:711–719. [PubMed] [Google Scholar]
- 142.Voehringer DW, McConkey DJ, McDonnell TJ, Brisbay S, Meyn RE. Bcl-2 expression causes redistribution of glutathione to the nucleus. Proc Natl Acad Sci U S A. 1998;95:2956–2960. doi: 10.1073/pnas.95.6.2956. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Amstad PA, Liu H, Ichimiya M, Berezesky IK, Trump BF, Buhimschi IA, et al. BCL-2 is involved in preventing oxidant-induced cell death and in decreasing oxygen radical production. Redox Rep. 2001;6(6):351–362. doi: 10.1179/135100001101536535. [DOI] [PubMed] [Google Scholar]
- 144.Ellerby LM, Ellerby HM, Park SM, Holleran AL, Murphy AN, Fiskum G, et al. Shift of the cellular oxidation-reduction potential in neural cells expressing Bcl-2. J Neurochem. 1996;67:1259–1267. doi: 10.1046/j.1471-4159.1996.67031259.x. [DOI] [PubMed] [Google Scholar]
- 145.Porcelli AM, Ghelli A, Iommarini L, Mariani E, Hoque M, Zanna C, et al. The antioxidant function of Bcl-2 preserves cytoskeletal stability of cells with defective respiratory complex I. Cell Mol Life Sci. 2008;65:2943–2951. doi: 10.1007/s00018-008-8300-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Zimmermann AK, Loucks FA, Schroeder EK, Bouchard RJ, Tyler KL, Linseman DA. Glutathione binding to the Bcl-2 homology-3 domain groove: a molecular basis for Bcl-2 antioxidant function at mitochondria. J Biol Chem. 2007;282:29296–29304. doi: 10.1074/jbc.M702853200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Mirkovic N, Voehringer DW, Story MD, McConkey DJ, McDonnell TJ, Meyn RE. Resistance to radiation-induced apoptosis in Bcl-2-expressing cells is reversed by depleting cellular thiols. Oncogene. 1997;15:1461–1470. doi: 10.1038/sj.onc.1201310. [DOI] [PubMed] [Google Scholar]
- 148.Voehringer DW, Meyn RE. Reversing drug resistance in bcl-2-expressing tumor cells by depleting glutathione. Drug Resist Updat. 1998;1(6):345–351. doi: 10.1016/s1368-7646(98)80010-1. [DOI] [PubMed] [Google Scholar]
- 149.Giardino I, Edelstein D, Brownlee M. BCL-2 expression or antioxidants prevent hyperglycemia-induced formation of intracellular advanced glycation endproducts in bovine endothelial cells. J Clin Invest. 1996;97:1422–1428. doi: 10.1172/JCI118563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Gilbert M, Knox S. Influence of Bcl-2 overexpression on Na+/K(+)-ATPase pump activity: correlation with radiation-induced programmed cell death. J Cell Physiol. 1997;171:299–304. doi: 10.1002/(SICI)1097-4652(199706)171:3<299::AID-JCP8>3.0.CO;2-J. [DOI] [PubMed] [Google Scholar]
- 151.Yin W, Cheng W, Shen W, Shu L, Zhao J, Zhang J, et al. Impairment of Na(+),K(+)-ATPase in CD95(APO-1)-induced human T-cell leukemia cell apoptosis mediated by glutathione depletion and generation of hydrogen peroxide. Leukemia. 2007;21:1669–1678. doi: 10.1038/sj.leu.2404791. [DOI] [PubMed] [Google Scholar]
- 152.Chen SR, Dunigan DD, Dickman MB. Bcl-2 family members inhibit oxidative stress-induced programmed cell death in Saccharomyces cerevisiae. Free Radic Biol Med. 2003;34:1315–1325. doi: 10.1016/s0891-5849(03)00146-1. [DOI] [PubMed] [Google Scholar]
- 153.Hochman A, Sternin H, Gorodin S, Korsmeyer S, Ziv I, Melamed E, et al. Enhanced oxidative stress and altered antioxidants in brains of Bcl-2-deficient mice. J Neurochem. 1998;71:741–748. doi: 10.1046/j.1471-4159.1998.71020741.x. [DOI] [PubMed] [Google Scholar]
- 154.Hochman A, Liang H, Offen D, Melamed E, Sternin H. Developmental changes in antioxidant enzymes and oxidative damage in kidneys,liver and brain of bcl-2 knockout mice. Cell Mol Biol (Noisy-le-grand) 2000;46:41–52. [PubMed] [Google Scholar]
- 155.Kondo S, Tang Y, Scheef EA, Sheibani N, Sorenson CM. Attenuation of retinal endothelial cell migration and capillary morphogenesis in the absence of bcl-2. Am J Physiol Cell Physiol. 2008;294:C1521–C1530. doi: 10.1152/ajpcell.90633.2007. [DOI] [PubMed] [Google Scholar]
- 156.Madeo F, Fröhlich E, Ligr M, Grey M, Sigrist SJ, Wolf DH, et al. Oxygen stress: a regulator of apoptosis in yeast. J Cell Biol. 1999;145:757–767. doi: 10.1083/jcb.145.4.757. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Nanbu-Wakao R, Asoh S, Nishimaki K, Tanaka R, Ohta S. Bacterial cell death induced by human pro-apoptotic Bax is blocked by an RNase E mutant that functions in an anti-oxidant pathway. Genes Cells. 2000;5:155–167. doi: 10.1046/j.1365-2443.2000.00316.x. [DOI] [PubMed] [Google Scholar]
- 158.Kampranis SC, Damianova R, Atallah M, Toby G, Kondi G, Tsichlis PN, et al. A novel plant glutathione S-transferase/peroxidase suppresses Bax lethality in yeast. J Biol Chem. 2000;275:29207–29216. doi: 10.1074/jbc.M002359200. [DOI] [PubMed] [Google Scholar]
- 159.Moon H, Baek D, Lee B, Prasad DT, Lee SY, Cho MJ, et al. Soybean ascorbate peroxidase suppresses Bax-induced apoptosis in yeast by inhibiting oxygen radical generation. Biochem Biophys Res Commun. 2002;290:457–462. doi: 10.1006/bbrc.2001.6208. [DOI] [PubMed] [Google Scholar]
- 160.Camougrand N, Grelaud-Coq A, Marza E, Priault M, Bessoule JJ, Manon S. The product of the UTH1 gene, required for Bax-induced cell death in yeast, is involved in the response to rapamycin. Mol Microbiol. 2003;47:495–506. doi: 10.1046/j.1365-2958.2003.03311.x. [DOI] [PubMed] [Google Scholar]
- 161.Chen S, Vaghchhipawala Z, Li W, Asard H, Dickman MB. Tomato phospholipid hydroperoxide glutathione peroxidase inhibits cell death induced by Bax and oxidative stresses in yeast and plants. Plant Physiol. 2004;135:1630–1641. doi: 10.1104/pp.103.038091. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Naderi J, Somayajulu-Nitu M, Mukerji A, Sharda P, Sikorska M, Borowy-Borowski H, et al. Water-soluble formulation of Coenzyme Q(10) inhibits Bax-induced destabilization of mitochondria in mammalian cells. Apoptosis. 2006;11:1359–1369. doi: 10.1007/s10495-006-8417-4. [DOI] [PubMed] [Google Scholar]
- 163.Wright SC, Wang H, Wei QS, Kinder DH, Larrick JW. Bcl-2-mediated resistance to apoptosis is associated with glutathione-induced inhibition of AP24 activation of nuclear DNA fragmentation. Cancer Res. 1998;58:5570–5576. [PubMed] [Google Scholar]
- 164.Armstrong JS, Jones DP. Glutathione depletion enforces the mitochondrial permeability transition and causes cell death in Bcl-2 overexpressing HL60 cells. FASEB J. 2002;16:1263–1265. doi: 10.1096/fj.02-0097fje. [DOI] [PubMed] [Google Scholar]
- 165.Steinman HM. The Bcl-2 oncoprotein functions as a pro-oxidant. J Biol Chem. 1995;270:3487–3490. [PubMed] [Google Scholar]
- 166.Papadopoulos MC, Koumenis IL, Xu L, Giffard RG. Potentiation of murine astrocyte antioxidant defence by bcl-2: protection in part reflects elevated glutathione levels. Eur J Neurosci. 1998;10:1252–1260. doi: 10.1046/j.1460-9568.1998.00134.x. [DOI] [PubMed] [Google Scholar]
- 167.Esposti MD, Hatzinisiriou I, McLennan H, Ralph S. Bcl-2 and mitochondrial oxygen radicals. New approaches with reactive oxygen species-sensitive probes. J Biol Chem. 1999 Oct 15;274(42):29831–29837. doi: 10.1074/jbc.274.42.29831. [DOI] [PubMed] [Google Scholar]
- 168.Kowaltowski AJ, Fenton RG, Fiskum G. Bcl-2 family proteins regulate mitochondrial reactive oxygen production and protect against oxidative stress. Free Radic Biol Med. 2004;37:1845–1853. doi: 10.1016/j.freeradbiomed.2004.09.005. [DOI] [PubMed] [Google Scholar]
- 169.López-Diazguerrero NE, López-Araiza H, Conde-Perezprina JC, Bucio L, Cárdenas-Aguayo MC, Ventura JL, et al. Bcl-2 protects against oxidative stress while inducing premature senescence. Free Radic Biol Med. 2006;40:1161–1169. doi: 10.1016/j.freeradbiomed.2005.11.002. [DOI] [PubMed] [Google Scholar]
- 170.Seyfried J, Evert BO, Schwarz CS, Schaupp M, Schulz JB, Klockgether T, et al. Gene dosage-dependent effects of bcl-2 expression on cellular survival and redox status. Free Radic Biol Med. 2003;34:1517–1530. doi: 10.1016/s0891-5849(03)00103-5. [DOI] [PubMed] [Google Scholar]
- 171.Hanson CJ, Bootman MD, Distelhorst CW, Maraldi T, Roderick HL. The cellular concentration of Bcl-2 determines its pro- or anti-apoptotic effect. Cell Calcium. 2008;44:243–258. doi: 10.1016/j.ceca.2007.11.014. [DOI] [PubMed] [Google Scholar]
- 172.Chen ZX, Pervaiz S. Bcl-2 induces pro-oxidant state by engaging mitochondrial respiration in tumor cells. Cell Death Differ. 2007;14:1617–1627. doi: 10.1038/sj.cdd.4402165. [DOI] [PubMed] [Google Scholar]
- 173.Ly JD, Grubb DR, Lawen A. The mitochondrial membrane potential (deltapsi(m)) in apoptosis; an update. Apoptosis. 2003;8:115–128. doi: 10.1023/a:1022945107762. [DOI] [PubMed] [Google Scholar]
- 174.Vander Heiden MG, Chandel NS, Williamson EK, Schumacker PT, Thompson CB. Bcl-xL regulates the membrane potential and volume homeostasis of mitochondria. Cell. 1997;91:627–637. doi: 10.1016/s0092-8674(00)80450-x. [DOI] [PubMed] [Google Scholar]
- 175.Vander Heiden MG, Chandel NS, Schumacker PT, Thompson CB. Bcl-xL prevents cell death following growth factor withdrawal by facilitating mitochondrial ATP/ADP exchange. Mol Cell. 1999 Feb;3(2):159–167. doi: 10.1016/s1097-2765(00)80307-x. [DOI] [PubMed] [Google Scholar]
- 176.Banki K, Hutter E, Gonchoroff NJ, Perl A. Elevation of mitochondrial transmembrane potential and reactive oxygen intermediate levels are early events and occur independently from activation of caspases in Fas signaling. J Immunol. 1999;162:1466–1479. [PMC free article] [PubMed] [Google Scholar]
- 177.Poot M, Pierce RH. Detection of changes in mitochondrial function during apoptosis by simultaneous staining with multiple fluorescent dyes and correlated multiparameter flow cytometry. Cytometry. 1999;35:311–317. doi: 10.1002/(sici)1097-0320(19990401)35:4<311::aid-cyto3>3.3.co;2-5. [DOI] [PubMed] [Google Scholar]
- 178.Krohn AJ, Wahlbrink T, Prehn JH. Mitochondrial depolarization is not required for neuronal apoptosis. J Neurosci. 1999;19:7394–7404. doi: 10.1523/JNEUROSCI.19-17-07394.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Scarlett JL, Sheard PW, Hughes G, Ledgerwood EC, Ku HH, Murphy MP. Changes in mitochondrial membrane potential during staurosporine-induced apoptosis in Jurkat cells. FEBS Lett. 2000;475:267–272. doi: 10.1016/s0014-5793(00)01681-1. [DOI] [PubMed] [Google Scholar]
- 180.Ouyang YB, Carriedo SG, Giffard RG. Effect of Bcl-x(L) overexpression on reactive oxygen species, intracellular calcium, and mitochondrial membrane potential following injury in astrocytes. Free Radic Biol Med. 2002;33:544–551. doi: 10.1016/s0891-5849(02)00912-7. [DOI] [PubMed] [Google Scholar]
- 181.Imahashi K, Schneider MD, Steenbergen C, Murphy E. Transgenic expression of Bcl-2 modulates energy metabolism, prevents cytosolic acidification during ischemia, and reduces ischemia/reperfusion injury. Circ Res. 2004;95:734–741. doi: 10.1161/01.RES.0000143898.67182.4c. [DOI] [PubMed] [Google Scholar]
- 182.Schwartz PS, Manion MK, Emerson CB, Fry JS, Schulz CM, Sweet IR, et al. 2-Methoxy antimycin reveals a unique mechanism for Bcl-x(L) inhibition. Mol Cancer Ther. 2007;6:2073–2080. doi: 10.1158/1535-7163.MCT-06-0767. [DOI] [PubMed] [Google Scholar]
- 183.Zhou YP, Pena JC, Roe MW, Mittal A, Levisetti M, Baldwin AC, et al. Overexpression of Bcl-x(L) in beta-cells prevents cell death but impairs mitochondrial signal for insulin secretion. Am J Physiol Endocrinol Metab. 2000;278:E340–E351. doi: 10.1152/ajpendo.2000.278.2.E340. [DOI] [PubMed] [Google Scholar]
- 184.Schwarz CS, Evert BO, Seyfried J, Schaupp M, Kunz WS, Vielhaber S, et al. Overexpression of bcl-2 results in reduction of cytochrome c content and inhibition of complex I activity. Biochem Biophys Res Commun. 2001;280:1021–1027. doi: 10.1006/bbrc.2001.4242. [DOI] [PubMed] [Google Scholar]
- 185.Vrbacký M, Krijt J, Drahota Z, Mĕlková Z. Inhibitory effects of Bcl-2 on mitochondrial respiration. Physiol Res. 2003;52:545–554. [PubMed] [Google Scholar]
- 186.Ricquier D, Bouillaud F. The uncoupling protein homologues: UCP1, UCP2, UCP3, StUCP and AtUCP. Biochem J. 2000;345:161–179. [PMC free article] [PubMed] [Google Scholar]
- 187.Wagner AM, Moore AL. Structure and function of the plant alternative oxidase: its putative role in the oxygen defence mechanism. Biosci Rep. 1997;17:319–333. doi: 10.1023/a:1027388729586. [DOI] [PubMed] [Google Scholar]