Skip to main content

Putting the pieces together: How is the mitochondrial pathway of apoptosis regulated in cancer and chemotherapy?

Abstract

In order to solve a jigsaw puzzle, one must first have the complete picture to logically connect the pieces. However, in cancer biology, we are still gaining an understanding of all the signaling pathways that promote tumorigenesis and how these pathways can be pharmacologically manipulated by conventional and targeted therapies. Despite not having complete knowledge of the mechanisms that cause cancer, the signaling networks responsible for cancer are becoming clearer, and this information is serving as a solid foundation for the development of rationally designed therapies. One goal of chemotherapy is to induce cancer cell death through the mitochondrial pathway of apoptosis. Within this review, we present the pathways that govern the cellular decision to undergo apoptosis as three distinct, yet connected puzzle pieces: (1) How do oncogene and tumor suppressor pathways regulate apoptosis upstream of mitochondria? (2) How does the B-cell lymphoma 2 (BCL-2) family influence tumorigenesis and chemotherapeutic responses? (3) How is post-mitochondrial outer membrane permeabilization (MOMP) regulation of cell death relevant in cancer? When these pieces are united, it is possible to appreciate how cancer signaling directly impacts upon the fundamental cellular mechanisms of apoptosis and potentially reveals novel pharmacological targets within these pathways that may enhance chemotherapeutic success.

Review

In multi-cellular organisms, cell growth, cell division, and cell death are regulated by a host of signaling pathways that integrate cellular condition and context. Within healthy tissues, there is a balance between these processes allowing for homeostasis. When this balance is perturbed, usually by uncontrolled proliferation and a collateral failure to activate cell death, susceptibility to cancer is increased. It has been suggested that there are as many ways to cause cancer as there are constellations in the sky—and we highlight a few of these pathways in our discussion below. Despite the many signaling pathways that lead to cancer vulnerability, most would agree that the best method to treat cancer is to specifically eliminate diseased cells via a genetically controlled program of cell death termed apoptosis.

Apoptosis is characterized by cysteine-aspartic protease (caspase)-dependent cleavage of numerous cellular substrates that allows for efficient packaging, detection, and elimination of the targeted cell from the surrounding environment. For our discussion, we will focus on the mitochondrial pathway of apoptosis, which means that mitochondria integrate the pro-apoptotic signaling environment via the B-cell lymphoma 2 (BCL-2) family of proteins to regulate cell death [1, 2]. The BCL-2 family controls the integrity of the outer mitochondrial membrane (OMM) and is functionally divided into anti- and pro-apoptotic proteins. Anti-apoptotic BCL-2 members (e.g., BCL-2/BCL-xL/MCL-1) preserve OMM integrity by directly sequestering the pro-apoptotic proteins, which cooperate to form pores with the OMM. Pore formation is referred to as mitochondrial outer membrane permeabilization (MOMP), and this results in the release of mitochondrial proteins (e.g., cytochrome c) that cooperate with cellular adaptor proteins (i.e., APAF-1) to induce caspase activation. From a mechanistic point of view, pro-apoptotic BCL-2 members are further divided into two subclasses: the effectors (e.g., BAX) and BH3-only proteins (e.g., BIM). It is suggested that BAX forms proteolipid pores within the OMM, and this process is nucleated by cooperative interactions with mitochondria and BH3-only proteins [3].

Returning to cancer-causing pathways, there are two general classes of proteins that promote tumorigenesis: oncogenes and tumor suppressors [4]. Oncogenic proteins normally function in homeostatic proliferative and survival mechanisms, but due to mutation (e.g., RASG12V and BRAFV600E) or divergent expression (e.g., BCL-2), these proteins undergo a gain of function to promote hyper-proliferation or sustained survival despite pro-apoptotic signaling. Likewise, tumor suppressor proteins (e.g., p53 and PTEN) negatively regulate survival and proliferation following cellular stress, but when mutated or deleted, they fail to appropriately restrain proliferation and this has potential to promote genomic instability and organelle dysfunction. In most cancers, distinct combinations of oncogenic signals and loss of tumor suppressor pathways drive tumorigenesis and resistance to apoptosis. Here, we will discuss specific examples of how oncogenic and tumor suppressor pathways intersect with the apoptotic machinery to alter apoptotic sensitivity, which ultimately impacts upon chemotherapeutic success and patient outcome.

Piece #1—How do oncogene and tumor suppressor pathways regulate apoptosis upstream of mitochondria?

Numerous oncogenic and tumor suppressor pathways converge on the BCL-2 family of proteins and downstream effectors to regulate cellular sensitivity to apoptosis during transformation as well as chemotherapeutic interventions. Here, we will discuss some of the most commonly altered tumor suppressor and oncogenic pathways that contribute to apoptotic defects in cancer (Figure 1). The tumor suppressor pathways (p53, retinoblastoma (Rb), and phosphatase and tensin homolog (PTEN)) and the proto-oncogene/oncogene pathways (Phosphoinositide 3-kinase/Akt (PI3K/AKT), RAS/RAF, Myc, and BCL-2) will be highlighted due to their broad implications in multiple tumor types [5, 6]. It is important to note that tumor suppressors and oncogenes function via transcriptional and/or non-transcriptional mechanisms. While the p53 tumor suppressor is an example of a protein that functions through both, other proteins such as BCL-2 function primarily through non-transcriptional means at the mitochondria, ER, and perhaps other cellular locations [1]. In the sections that follow, we will discuss these pathways in more detail.

Figure 1
figure 1

Piece #1: Tumor suppressor and oncogenic pathways converge on the mitochondrial pathway of apoptosis. Oncogenic (e.g., PI3K/AKT, RAS-MAPK, and Myc) and tumor suppressor pathways (e.g., p53, PTEN, and Rb) act at transcriptional and non-transcriptional levels to modulate cellular sensitivity to detect and repair stress, along with regulating the expression and function of downstream apoptotic proteins. Details are provided in the text.

What is the role of the tumor suppressor network in apoptosis?

One of the major regulators of apoptotic signaling following oncogenic (e.g., aberrant Myc expression) and pharmacological stress (e.g., conventional chemotherapy) is the p53 pathway. p53, often referred to as “guardian of the genome”, is a transcription factor that regulates cellular responses to a multitude of stresses including DNA damage, oncogene activation, and cell cycle and metabolic aberrations [7, 8]. In the event of acute stress, the p53 pathway ensures that DNA damage events are allowed to repair prior to mitosis [911]. However, if stress is chronic and/or repair mechanisms insufficient, pro-apoptotic signaling mediated by p53 acts to eliminate the affected cell [7].

To commit a cell to apoptosis, p53 acts through both transcriptional and non-transcriptional mechanisms. p53 sensitizes cells to apoptosis through direct transcriptional induction of numerous pro-apoptotic members of the BCL-2 family including, BAX, Noxa, and PUMA[1215]. In addition to its transcriptional role, p53 directly interacts with multiple members of the BCL-2 family to regulate MOMP. For example, cytosolic and mitochondrial forms of p53 have been shown to directly activate the pro-apoptotic effectors BAK/BAX as well as bind and inhibit the anti-apoptotic proteins BCL-xL and BCL-2 [1619]. The integration of p53 at multiple points in the mitochondrial pathway of apoptosis highlights the crucial role for this tumor suppressor pathway in the cellular decision to commit to apoptosis.

Another tumor suppressor with highly aberrant expression in many cancers is the Rb protein [2024]. In unstressed cells, Rb is generally maintained at a hypo-phosphorylated state, which favors a Rb-E2F interaction. During G1, hyper-phosphorylation of Rb by CDK/cyclin complexes disrupts this interaction, thereby de-repressing E2F and allowing for the transcription of genes required for cell cycle progression [25]. Over the years, however, additional anti- as well as pro-survival roles have been described for Rb [2632]. Consistent with a tumor suppressive function, a pro-apoptotic role for Rb has been described in studies using various cancer cell lines, including glioblastoma, prostate, and cervical cancers [2729]. In this context, Rb was shown to induce apoptosis in response to genotoxic and oncogenic stresses by promoting transcriptional activation of pro-apoptotic proteins [33]. More recently, Rb was reported to localize to mitochondria and induce apoptosis through direct activation of BAX [34, 35]. Interestingly, an anti-apoptotic role has also been described for the protein. Rb has been shown to decrease apoptotic sensitivity in mouse cell lines (again through E2F1 repression) by lowering expression levels of APAF-1 and caspase-9 [31, 36, 37]. These opposing functions suggest a context-dependent role for Rb in the regulation of apoptosis.

Tumor suppressors are considered the “sentinels” of a cell that protect from oncogenic aberrations to restrict proliferation to healthy cells. Moreover, these pathways function to detect oncogenic stress and/or DNA damage to halt proliferation. It is for these reasons that pre-malignant cells select against this first line of defense in order to initiate tumorigenesis.

How does oncogenic signaling regulate apoptosis?

A common driver of oncogenesis is the alteration of genes through mutation or chromosomal aberration. While proto-oncogenes ensure a balance between survival and apoptosis to maintain healthy tissues, their mutant form, oncogenes, shifts this balance to favor cell survival, proliferation, and resistance to cell death.

The PI3K/AKT pathway plays a major role in promoting many tumor types. PI3K/AKT is among the most frequently mutated network in cancer [38, 39], which leads to massive hyper-activation of this potent survival and proliferation pathway. In addition, several cancers reduce the negative regulator of the pathway, PTEN, a commonly mutated tumor suppressor. PTEN is a dual specificity protein and lipid phosphatase that localizes mainly to the cytosol but is suggested to function in the nucleus and extracellular matrix [40, 41]. PTEN negatively regulates the PI3K/AKT pathway by inhibiting the PIP3-dependent activation of AKT [42]. Once active, AKT phosphorylates numerous downstream substrates, including transcription factors as well as direct regulators of apoptosis. Examples of these include the FOXO family of transcription factors which are phosphorylated and inactivated by AKT, resulting in decreased expression of their target pro-apoptotic proteins BIM and PUMA[4345]. In addition, AKT directly phosphorylates and suppresses the function of the pro-apoptotic BCL-2 family proteins BAD, BIM, and BAX and upregulates the levels of X-linked inhibitor of apoptosis protein (XIAP) through increased protein stability [43, 4648]. Taken together, the activating mutations in the PI3K/AKT pathway, combined with the inactivation of the PTEN tumor suppressor, result in oncogenic activation of one of the most formidable signaling pathways in cancer. Targeting this pathway at tumor suppressor (i.e., PTEN) and oncogene levels gives the advantage of not only attacking the pro-survival arm of the pathway but also ensuring apoptosis induction through restoration of its tumor suppressor function as well.

The RAS/mitogen-activated protein kinase (MAPK) pathway is another major cellular signaling network that commonly acquires oncogenic mutations at various points in the pathway. For example, mutations in receptor tyrosine kinases (e.g., EGFR, ErbB2), the small GTPase RAS (i.e., RASG12V), and downstream RAF kinases (e.g., BRAFV600E) are described in a variety of cancers [38, 4953]. The pathway proceeds via a series of intermediate kinases leading to the activation of extracellular receptor kinase (ERK), which regulates the transcriptional activation of many genes involved in cell cycle and apoptosis. ERK signaling has been shown to transcriptionally activate the pro-survival genes BCL-2 and BCL-xL, as well as stabilize MCL-1 through phosphorylation [54, 55]. It has been reported that oncogenic ERK activation leads to a decrease in expression levels of BIM, as well as proteasomal degradation of BIM through direct phosphorylation of the protein [5658]; all of which can be reversed by small molecule inhibition of the pathway [5962]. In addition, kinases downstream of ERK (e.g., RSK, S6K) directly phosphorylate and inactivate the pro-apoptotic BCL-2 family member BAD, as well as caspase-9 and APAF-1 [6366].

Myc is a classic oncogenic transcription factor that is over-expressed in a large number of human cancers. Myc expression is upregulated through a variety of mechanisms including chromosomal translocations and amplifications, activation of upstream growth signaling pathways, and increased protein stability [67]. Myc was one of the first proteins identified to have antagonistic pleiotropic functions, promoting both cell survival and cell death [68]. The paradox arises from the oncogene’s ability to cause apoptosis when over expressed, and it has been suggested that this apoptotic phenotype is a measure to ensure protection against unrestricted proliferation, and is bypassed during tumorigenesis [67]. Myc-induced apoptosis can be p53 dependent based on cell type and apoptotic stimulus. Upregulation of p53 by Myc increases the expression of the pro-apoptotic BCL-2 family members, BAX, PUMA, and Noxa[1215]. Alternatively, a p53-independent mechanism of Myc exists by either directly suppressing BCL-2 expression in a cell type-specific manner or directly acting on BIM expression [69]. More recently, the oncometabolite 2-hydroxyglutarate from isocitrate dehydrogenase mutant cancers was found to directly activate Myc-mediated apoptosis in breast cancer [70], suggesting that Myc may be an important link between altered cellular metabolism and apoptosis in cancer.

The focus of this section thus far has been on how potent oncogenes function to ensure cell survival and target apoptotic pathways to reduce cell death sensitivity. Last but not least on this list comes the founding member of the BCL-2 family itself. Originally identified as a chromosomal translocation in B-cell lymphoma, BCL-2 is the founding member of the family that is responsible for directly inhibiting the mitochondrial pathway of apoptosis [71]. The translocation identified in B-cell lymphoma positions BCL-2 under the control of the immunoglobulin heavy-chain promoter, leading to massive BCL-2 over-expression and subsequent resistance to cell death. The function of BCL-2 as an oncogene is unusual in that over-expression alone is not sufficient to drive cellular transformation but requires additional oncogenes (e.g., Myc) [72]. This result revealed that BCL-2 does not promote cell proliferation, but rather it blocks pro-apoptotic signals from collateral oncogenes. While the example of BCL-2 translocation in lymphoma is not observed in many tumor types, over-expression of anti-apoptotic members of the BCL-2 family is a common feature in cancers of the uterus, lung, ovary, breast, colon, liver, and gastrointestinal tract [7376]. The mechanism by which BCL-2 expression directly controls apoptosis will be discussed shortly.

The oncogenic and tumor suppressor pathways mutated in cancer have become major targets for drug development over the past few decades. While most conventional chemotherapy responses proceed via the mitochondrial pathway of apoptosis (often mediated by DNA damage and p53), more recently there has been explicit focus on the development of targeted therapies for specific proteins within these tumorigenic pathways. Table 1 presents a sampling of the current and developing drugs targeting the tumor suppressors and oncogenes described above. While tumor suppressor and oncogenic pathways require mitochondrial contributions to die, the cellular decision to initiate MOMP and apoptosis is governed by the functional repertoire of BCL-2 family proteins at the OMM. In the next section, we will discuss how the BCL-2 family of proteins impacts upon the execution of the mitochondrial pathway of apoptosis in response cancer cell signaling and chemotherapeutics.

Table 1 Drugs currently in clinical trials targeting tumor suppressor/oncogene pathways or proteins within the mitochondrial pathway of apoptosis

Piece #2—How does the BCL-2 family influence tumorigenesis and chemotherapeutic responses?

The BCL-2 family comprises four groups of proteins, which are based on their domain composition (i.e., one to four BCL-2 homology “BH” domains) and function [77]. Pro-apoptotic effectors (BAK and BAX) are mechanistically involved in MOMP as these proteins interact with the OMM, leading to its permeabilization [77]. In order for BAK or BAX to initiate MOMP, they must be localized to the OMM, which creates the appropriate biophysical and biochemical environment to support structural rearrangements that nucleate homo-oligomerization into proteolipid pores [78, 79]. The structural rearrangements can be triggered by various mechanisms, but the one most relevant to our discussion is via protein-protein interactions with the direct activator BH3-only proteins (e.g., BID and BIM) [8082]. Through direct binding of these proteins to either the N-terminus of BAK/BAX or a hydrophobic region in the core of the protein formed by the BH1–3 domains, the direct activators trigger essential conformational changes that render these effector proteins competent to oligomerize [8284]. Conversely, the anti-apoptotic proteins (e.g., BCL-2, BCL-xL/BCL2L1, BCL-w/BCL2L2, A1/BCL2A1, and MCL-1) inhibit BAK/BAX activation by direct interaction or by sequestration of the direct activator BH3-only proteins [80, 81]. This inhibition can be prevented (termed “sensitization”) or displaced (termed “de-repression”) by the final group of BCL-2 family members, the sensitizer/de-repressor BH3-only proteins (e.g., BAD, Noxa, and PUMA) [80, 85]. As the role of the BH3-only proteins is significant in the regulation of cellular sensitivity to apoptosis via BAK and BAX activation, we will discuss several approaches to regulate BH3-only protein function by intracellular signaling pathways and small molecules that were designed to mimic their action. The mechanism of action of the BCL-2 family to regulate apoptosis is summarized in Figure 2.

Figure 2
figure 2

Piece #2: The BCL-2 family controls BAK/BAX activation and MOMP. Pro-apoptotic BCL-2 family protein activation is triggered by extra- and intra-cellular signaling. De-repressor BH3-only proteins (green) prevent or disrupt inhibition by anti-apoptotic proteins (red). Direct activator BH3-only proteins (yellow) bind BAK and BAX (blue) to induce their homo-oligomerization and MOMP. Details are provided in the text.

BCL-2 family deregulation in cancer

The regulation of MOMP is complex due to multiple proteins and pathways converging upon the BCL-2 family; furthermore, there are specific expression and functional patterns that are dependent upon cell type and differentiation state [86]. What is key to understanding how the BCL-2 family regulates apoptosis in cancer is directly linked to the mechanisms described earlier, those being sensitization, de-repression, and direct activation of BAK/BAX. In addition to the above BCL-2 translocation event, epigenetic regulation of anti-apoptotic BCL-2 proteins also plays a role in reducing cellular sensitivity to apoptosis. As an example, hypo-methylation of the BCL-2 promoter has been reported in chronic lymphocytic leukemia (CLL) [87]. Of course, the expression of anti-apoptotic proteins is positively selected during transformation because the targeted cell is trying to eliminate itself through pro-apoptotic signaling, yet oncogenic and tumor suppressor pathways must promote anti-apoptotic BCL-2 family function to survive [88]. The dual upregulation of pro-apoptotic and anti-apoptotic proteins is referred to as “priming”, which means the cells are uniquely poised to engage apoptosis due to constitutive sequestration of pro-apoptotic proteins, such as BIM. The presence of sequestered BIM presents a pharmacological opportunity to treat primed cancer cells with BH3 mimetics (discussed below) as pro-apoptotic signaling appears intact [89, 90].

Post-transcriptionally, several cancer-associated miRNAs are involved in the control of the BCL-2 family. For example, miR-15a and miR-16-1 are reduced in about two thirds of B-cell CLL cases resulting in BCL-2 over-expression and the establishment of disease [91]. Other miRNAs in CLL, such as miR-181a/b, attenuate BCL-2 and BCL-xL expression and are markers of chemotherapeutic success [92]. In addition to regulation at the transcriptional and translational levels, members of the BCL-2 family are controlled by a variety of post-translational modifications. For instance, BAD phosphorylation on serines 112 and 136 is exacerbated in glioblastomas, prostate cancers, and melanomas due to a combination of oncogenic MAPK signaling and PTEN mutation/downregulation [93]. This situation likely mediates sensitivity to apoptosis by altering the affinity of BAD for anti-apoptotic partners, thereby influencing sensitization and de-repression mechanisms. On a similar note, BIM-EL (one of three BIM isoforms) phosphorylation at serine 69 by oncogenic MAPK signaling influences associations with MCL-1 and correlates with resistance to apoptosis in CLL [94]. Altogether, the above examples show that the BCL-2 family proteins are regulated at the genomic, translational, and post-translational levels by cancer-associated pathways.

How do we pharmacologically target the BCL-2 family?

In order to engage apoptosis, BH3-only proteins must interact with anti- and pro-apoptotic BCL-2 proteins; therefore, the majority of small molecules identified to regulate apoptotic sensitivity mimic these interactions. One of the first natural BH3-mimetic molecules discovered was gossypol, a polyphenol extracted from cottonseed [95]. Gossypol and its derivative, TW-37, and apogossypolone (ApoG2) target BCL-2, BCL-xL, and MCL-1, and effectively promote apoptosis in lung, prostate, and lymphoma cancer models [9698]. In parallel to naturally derived compounds, numerous small molecules were engineered through structure activity relationship strategies to target the hydrophobic groove of anti-apoptotic BCL-2 proteins. For example, chemical engineering and assembly of several low affinity molecules led to the generation of the highly specific drug ABT-737 [99]. Despite lacking some key pharmacological properties required to be used in the clinic (e.g., non orally bio-available), the discovery of ABT-737 constituted a milestone in specifically targeting the BCL-2 family, and further modification of this drug led to the bioavailable derivative, ABT-263. ABT-737 and ABT-263 are highly specific for BCL-2, BCL-xL, and BCL-w and have proven efficacy on BCL-2/BCL-xL-dependent tumors such as leukemia and lymphoma [99]. As an aside, one commonly observed side effect of ABT-263 therapy is rapid thrombocytopenia, which occurs because platelets rely exclusively on BCL-xL for survival [100, 101]. To avoid this phenotype, an additional derivative (ABT-199) was generated that has markedly reduced its affinity for BCL-xL, and is therefore more specific to BCL-2 [102]. Indeed, ABT-199 was shown to retain the same efficiency as ABT-737 on leukemia and lymphomas without the collateral thrombocytopenia [102, 103].

Consistent with the development of rational drug design to target anti-apoptotic BCL-2 proteins, new molecules were recently reported, including MIM1 [104], Terphenyl-14, and WEHI-539 [105, 106], which specifically target MCL-1 and BCL-xL, respectively. These pharmacological agents are highly significant to designing precision treatments as tumors frequently display dependency upon BCL-2 or MCL-1, and chemo-resistant tumors often shift reliance between anti-apoptotic BCL-2 proteins. Importantly, the dependency upon different anti-apoptotic BCL-2 proteins can be determined by BH3 profiling to reveal which patients are most likely to respond to conventional chemotherapy [107, 108]. Interestingly, recent evidence suggests that response to BH-3 mimetics is not only determined by the anti-apoptotic proteins, but the pro-apoptotic repertoire as well [109]. In addition to targeting anti-apoptotic BCL-2 members, recent BH3-mimetics design has generated small molecules that function similar to direct activator BH3-only proteins to directly induce BAX activation and MOMP. For example, the small molecule BAX activator molecule 7 (BAM7) demonstrates impressive potency to activate BAX, similar to BIM in transformed cells [110]. Of course, one relevant question is how will this novel class of molecules be used to specifically kill cancer cells? It is likely that novel combinations of sub-threshold levels of chemotherapeutics will provide the best patient benefits.

As discussed earlier, BH3 mimetics are useful as single agents in hematological malignancies harboring BIM; however, the majority of solid tumors do not constitutively express direct activator BH3-only proteins [111]. Therefore, the design of combination therapies must incorporate strategies to induce direct activator BH3-only protein expression in order to sensitize solid tumors to BH3 mimetics. For example, inhibition of BRAFV600E signaling by PLX-4032 triggers a stress response that leads to increased expression and accumulation of BIM at the OMM [61, 112]. These sequestered molecules of BIM can be functionalized by the collateral inhibition of the anti-apoptotic BCL-2 repertoire using ABT-737 for example [61]. Similar approaches using conventional chemotherapies have generated comparable results, suggesting broad applications for these therapeutic strategies [113].

Piece #3—How is post-MOMP regulation of cell death relevant in cancer?

So far, we discussed the various cancer-related signaling pathways upstream of mitochondria taking into consideration the dynamic interactions within the BCL-2 family at the OMM that lead to the decision to die. Following MOMP, a cell normally enters the final stages of demise; while this is often considered the “point of no return”, there is a growing literature suggesting cells maintain the ability to resist cell death despite caspase activation [114]. Here, we will highlight several cellular mechanisms that regulate cell fate post-MOMP including intermembrane space (IMS) protein release, caspase activation, and cellular clearance. The therapeutic opportunities to target these final stages of apoptosis will also be discussed.

What happens post-MOMP?

Once BAK/BAX homo-oligomers permeabilize the OMM, the inner mitochondrial membrane (IMM) and cristae junctions undergo extensive remodeling [115117]. This remodeling allows several IMS proteins to diffuse into the cytosol (e.g., cytochrome c, SMAC, Omi, and nearly all soluble IMS proteins). One of the most crucial of these proteins is cytochrome c, which binds the cytosolic adaptor protein apoptotic protease activating factor (APAF-1) and triggers the formation of a heptameric complex that recruits and activates procaspase-9. Activated dimeric caspase-9 directly cleaves and activates the downstream effector caspase-3 and caspase-7 (Figure 3). These effector caspases are responsible for eliciting the classical apoptotic phenotypes characterized by DNA laddering, phosphatidylserine exposure, and contraction from the surrounding healthy cells. The cellular phenotypes stemming from caspase activation also allow for the generation of “find me” and “eat me” signals that elicit phagocytosis and removal of the dying cell [118, 119].

Figure 3
figure 3

Piece #3: Post-MOMP regulation of cell death. Pro-apoptotic proteins within the mitochondrial inter-membrane space (e.g., cytochrome c, SMAC, and Omi) are released after MOMP and directly regulate the activation of caspases and commitment to apoptosis. Details are provided in the text.

As we deepen our mechanistic understanding of how the mitochondrial pathway of apoptosis proceeds after MOMP, the majority of the literature would agree with the notion that irrespective of caspase activation (i.e., caspase activation promotes rapid packaging and detection, but the inhibition of caspases will only delay, not prevent cell death), most cells die following MOMP due to aberrations in mitochondrial biology. However, the general applicability of this concept is increasingly being called into question. Genetic evidence unquestionably supports a pro-survival role for cytochrome c as an integral part of the electron transport chain; cytochrome c knockout mice are embryonic lethal due to an organism-wide failure to generate ATP, and tissue-specific deletions of cytochrome c corroborate these results [120, 121]. Somewhat paradoxically, cytochrome c also functions as a crucial mediator of caspase-dependent death; cells deficient in cytochrome c are resistant to cytotoxic insults [122]. Whole organism or tissue-specific deletion eliminates cellular and/or tissue viability, presumably through a reduction in ATP generation and developmental cell death that is required for tissue and organ function. Interestingly, developmental phenotypes are shared with downstream apoptotic counterparts. Apaf1 and Caspase9 deficiencies result in the inhibition of developmental apoptosis, with phenotypes most usually characterized by exencephaly and cranioschisis [123, 124]. The function of these proteins in tumor suppression however remains controversial. While genetic studies have shown that Apaf-1 and/or caspase-9 deletion promote Myc-induced oncogenic transformation of MEFs, in vivo deletion of these genes reportedly had no effect on the rate, severity, or chemotherapeutic response of Myc-induced lymphomas [125, 126]. This is in contrast to what is observed, for example, with deletions of pro-apoptotic proteins such as BIM and BAD. Deletions of either of these genes have been shown to enhance Eμ-myc induced lymphoma, highlighting their importance in suppressing lymphomagenesis [127, 128].

To ensure that pro-apoptotic caspases are not inappropriately activated in unstressed cellular conditions, additional “apoptotic brakes” are in place that prevent caspase activation [129]. One example is XIAP, which promotes cellular survival by inhibiting caspase activation via direct protein-protein interactions [130]. Following MOMP, the anti-apoptotic activity of XIAP is counteracted by the release of two IMS proteins: second mitochondria-derived activator of caspase (SMAC) and Omi/Htra2 (Omi). Once released into the cytosol, SMAC and Omi bind and antagonize the activity of XIAP, thereby allowing for caspase activation to proceed [131, 132]. The function of SMAC and Omi suggests that post-MOMP regulation of caspase activity is required, which would not be the case if MOMP was always sufficient to promote death. It is important to mention that the Smac and Omi knockout mice develop normally and exhibit no defects in susceptibility to apoptosis [133]. This suggests a possible redundancy in the function of these proteins or a specificity in cellular stress conditions. Despite an unclear role in apoptosis, both of these proteins also have been suggested to play a role in cancer progression and chemotherapeutic responses. A decrease in SMAC expression, at the mRNA and protein levels, has been reported in many malignancies including renal cell carcinoma, hepatocellular carcinoma, testicular cancer, and lung cancer. Interestingly, in many of these studies, a decrease in SMAC levels was also accompanied by an increase in inhibitor of apoptosis protein (IAP) expression level as well as an increase in tumor invasion and metastasis [134138].

It has become evident that in many cell types, there is an anti-apoptotic threshold for endogenous caspase activation, as well as XIAP levels, to modulate cell death responses. This notion is supported by the observation that irradiation of cells leading to permeabilization of up to 15% of the mitochondrial population does not induce an apoptotic response, suggesting that local release of mitochondrial proteins does not result in an amplifiable apoptotic signal [139]. This may also explain the contribution of XIAP over-expression in many tumors, thereby increasing the threshold for caspase activation and efficient execution of cell death. It is worth mentioning that XIAP knockout mice are viable and lack apoptotic defects. These mice do, however, show increases in cellular IAP (c-IAPs) protein levels suggesting that these proteins may compensate for XIAP loss during development and apoptosis [140].

The ultimate goal of post mitochondrial regulation of pro-apoptotic BCL-2 family function and MOMP is to initiate the activation of caspases that will complete the apoptotic program. It is important to consider however that while caspases play a role in mediating cell death, they also play important roles in maintaining cell survival. Caspases generally thought to function exclusively in apoptosis are now being reported to have many additional cellular functions [141]. Executioner caspases have been shown to play roles in adaptive immunity as well as cell fate decisions including cell differentiation and migration [142145]. This raises the question of how a cell can differentiate between apoptotic and non-apoptotic caspase activation. Studies have suggested that a threshold of caspase activation exists in cells where only small levels of activation are required for non-apoptotic functions, whereas much higher levels are required to execute cell death. Another possible mechanism of regulated non-apoptotic caspase activation is the compartmentalization of active caspases. Examples of such mechanisms have been demonstrated in neurons, as well as in macrophages where caspase containing inflammasomes have been shown to form. In cancer, overall levels of caspases, particularly executioner caspases, can be expressed at very low levels. A screen of primary breast tumors found that approximately 75% of tumors lacked CASP3 transcript as well as protein expression [146], and similar findings were reported in colorectal and gastric tumors, which were found to express very low or absent levels of caspase-7 [147, 148]. It is important to mention however that due to the redundancy of these proteins, very little evidence supports a role for individual caspases in regulating tumorigenesis. Individual caspase knockout animals exhibit quite mild phenotypes and cells derived from these mice are only slightly more resistant to apoptosis than their WT counterparts. Cells lacking both CASP3 and CASP7 however are extremely resistant to apoptotic stimulus [149]. These observations raise the possibility that low levels of caspase activation may promote cell survival and/or tumorigenesis. Among the demonstrated non-apoptotic roles of caspases is role in cell migration and potentially invasiveness [150, 151]. It is possible that low or basal levels of caspases promote cellular migration to a more tumor favorable milieu.

Is there regulation of apoptosis after MOMP?

Given the indication that several mechanisms are in place to regulate caspase activation and apoptosis post-MOMP, the next question that arises is why a cell would need to commit resources to do so once mitochondrial integrity has been compromised. As previously mentioned, it appears that a specific threshold of cytochrome c release and subsequent caspase activation must be reached in order to elicit an apoptotic response. This may be a mechanism to ensure that a cell survives any potential “accidental MOMP” events. Recovery post-MOMP may also be essential for post-mitotic cells including cardiomyocytes and sympathetic neurons. Such tissues exhibit poor regenerative potential and therefore have adapted mechanisms to ensure longevity despite incomplete MOMP [152, 153]. Lower APAF-1 levels have been reported in both cell types as well as resistance to cytochrome c microinjection. Inhibition of XIAP through the addition of recombinant SMAC or deletion of XIAP resensitizes these cells, which further highlights the importance of XIAP in maintaining cellular survival [154156].

Finally, ensuring regulation of cell death post-MOMP is essential for recovery in proliferating cells and has important implications for tumorigenesis. As discussed throughout this section, tumors have been shown to develop mechanisms such as loss of APAF-1, defective caspase activation, and upregulation of XIAP to bypass complete cell death [157161]. Cancer-associated pathways like PI3K/AKT have been shown to antagonize caspase activity by phosphorylation of caspase-9 and caspase-3 [151, 162]. The cellular mechanisms related to caspase inhibition post-MOMP may present interesting therapeutic opportunities that can be exploited for cancer treatment.

Can cells survive despite MOMP?

As discussed, cytochrome c is not only essential for apoptosome formation but is also an essential component of the electron transport chain. Once MOMP has occurred and cytochrome c is released, not only does this trigger the apoptotic cascade but also transiently shuts down the electron transport chain. One would expect that both these events would effectively render cell survival post MOMP unlikely; however, there is evidence of scenarios where cells do recover and survive. This paradox raises the question of how cells can survive once MOMP has occurred. Interestingly, a study by Colell et al. implicated Glyceraldehyde 3-phosphate dehydrogenase (GAPDH) in mediating cellular recovery following MOMP. The authors showed that through enhanced glycolysis and autophagy, GAPDH could mediate clonogenic survival post-MOMP if caspase activation was inhibited [163]. In addition, work by Tait et al., in 2010 demonstrated that often, cells undergo incomplete MOMP. Through live cell imaging, it was determined that not all mitochondria in a cell undergo MOMP in response to apoptotic stimulus. The small surviving population provides a cohort of intact, healthy mitochondria that can potentially repopulate the mitochondrial network and allow for full cell recovery [164]. Not only do these studies demonstrate how cells could potentially survive once MOMP has occurred but also they further underscore the importance of caspases in mediating apoptosis. While these studies propose interesting mechanisms of post-MOMP recovery of cells, the significance of these processes has yet to be explored in a tumorigenic setting.

How can post-MOMP events be targeted for therapeutic purposes?

The majority of cell death research with direct implications on killing cancer cells has focused on the identification of pathways and therapeutics that promote apoptosis at the levels of pro-apoptotic signaling (Piece #1) and the BCL-2 family (Piece #2). Given that many tumors have adapted mechanisms to reduce apoptosis by regulating activities following MOMP, targeting post-mitochondrial proteins may present novel therapeutic opportunities.

In 2000, the first crystal structure of the interaction between SMAC and IAPs was reported [165167]. This structure served as the basis for the development of SMAC mimetics to act as IAP antagonists. These peptides have been shown to effectively inhibit IAP activity in several cancer cells, thereby sensitizing them to pro-apoptotic stimuli [168]. In non-small cell lung cancer, SMAC mimetic JP1201 was shown to sensitize cells to standard chemotherapy [169]. The same peptide was also shown to reduce primary and metastatic tumor burden in xenograft models of pancreatic cancer when used in combination with chemotherapeutics [170]. Interestingly, not only do these molecules sensitize cells to mitochondrial apoptosis through XIAP degradation but also to TNF-induced cell death by antagonizing cellular IAPs. Indeed, SMAC mimetics can sensitize to inducers of non-apoptotic cell death via the regulation of TNF receptor mediated signaling, and this is also influenced by pro-survival pathways, such as NFκB [171]. Several other SMAC mimetics have also been developed and are beginning to show efficacy in phase I and II clinical trials (see Table 1). In addition to SMAC mimetics, several IAP antagonists have been developed, including specific XIAP and cIAP inhibitors as well as XIAP antisense oligonucleotides. The latter has shown promising effects in phase I and II clinical trials when used in combination with standard chemotherapy in patients with acute myeloid leukemia [172].

Conclusions

The focus of our discussion has been to describe the numerous mechanisms by which tumor suppressor and oncogenic pathways reduce apoptotic sensitivity to initiate tumorigenesis and how these aberrations ultimately impact upon the success of chemotherapeutic interventions. From the evidence provided above, it appears that there are two pro-apoptotic signaling networks that may be specifically disrupted to ensure the survival of cells harboring oncogenic signals (e.g., oncogenic MAPK signaling) or genomic instability (e.g., DNA lesions). The first being upstream of the core apoptotic machinery; this includes the proteins and pathways (e.g., the p53 pathway) that specifically detect and respond to oncogenic signaling and macromolecular damage. When these pathways fail to recognize aberrations, the compromised cell does not initiate cell cycle arrest and repair mechanisms to maintain stability. In situations of chronic or irreparable cellular stress, a cell may be able to detect cellular damage, but if the pro-apoptotic machinery is not effectively engaged to eliminate the compromised cell (e.g., BCL-2 over-expression), its persistence increases the likelihood of developing and maintaining secondary events that may initiate malignancy, and potentially, chemotherapeutically intractable disease.

Since the advent of cancer chemotherapy, conventional treatments that promote apoptosis (e.g., cisplatin, dacarbazine, vinblastine) have provided the bulk of positive patient responses and remissions, yet the negative side effects and low response rates for many tumor types force scientists and clinicians to search for more optimal strategies. Given our broader knowledge of how the above pathways function in both physiological and pathophysiological apoptosis, it is being increasingly evident that pharmacologically targeting the specific upstream (e.g., BRAFV600E) and/or direct pro-apoptotic signaling pathways (e.g., BH3 mimetics) will likely provide a patient benefit. Returning to the jigsaw puzzle analogy mentioned earlier, our discussion on the three key distinct steps (or puzzle pieces) that regulate apoptotic sensitivity before and after chemotherapeutic interventions reveals that we are making significant progress in understanding the key contributions of apoptosis in cancer and chemotherapy. Likewise, as we continue to identify mutations and mechanisms that directly control apoptosis and malignancy, our pharmacological space to rationally design small molecules will hopefully allow for enhanced precision medicine to specifically eradicate malignant cells.

References

  1. Chipuk JE, Moldoveanu T, Llambi F, Parsons MJ, Green DR: The BCL-2 family reunion. Mol Cell. 2010, 37 (3): 299-310. 10.1016/j.molcel.2010.01.025.

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Tait SW, Green DR: Mitochondrial regulation of cell death. Cold Spring Harb Perspect Biol. 2013, 5 (9):

  3. Walensky LD, Gavathiotis E: BAX unleashed: the biochemical transformation of an inactive cytosolic monomer into a toxic mitochondrial pore. Trends Biochem Sci. 2011, 36 (12): 642-652. 10.1016/j.tibs.2011.08.009.

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Hanahan D, Weinberg RA: Hallmarks of cancer: the next generation. Cell. 2011, 144 (5): 646-674. 10.1016/j.cell.2011.02.013.

    CAS  PubMed  Google Scholar 

  5. Delbridge AR, Valente LJ, Strasser A: The role of the apoptotic machinery in tumor suppression. Cold Spring Harb Perspect Biol. 2012, 4 (11):

  6. Shortt J, Johnstone RW: Oncogenes in cell survival and cell death. Cold Spring Harb Perspect Biol. 2012, 4 (12):

  7. Vousden KH, Prives C: Blinded by the light: the growing complexity of p53. Cell. 2009, 137 (3): 413-431. 10.1016/j.cell.2009.04.037.

    CAS  PubMed  Google Scholar 

  8. Vousden KH, Lane DP: p53 in health and disease. Nat Rev Mol Cell Biol. 2007, 8 (4): 275-283. 10.1038/nrm2147.

    CAS  PubMed  Google Scholar 

  9. Vogelstein B, Lane D, Levine AJ: Surfing the p53 network. Nature. 2000, 408 (6810): 307-310. 10.1038/35042675.

    CAS  PubMed  Google Scholar 

  10. Sengupta S, Harris CC: p53: traffic cop at the crossroads of DNA repair and recombination. Nat Rev Mol Cell Biol. 2005, 6 (1): 44-55. 10.1038/nrm1546.

    CAS  PubMed  Google Scholar 

  11. Mirza A, Wu Q, Wang L, McClanahan T, Bishop WR, Gheyas F, Ding W, Hutchins B, Hockenberry T, Kirschmeier P, Greene JR, Liu S: Global transcriptional program of p53 target genes during the process of apoptosis and cell cycle progression. Oncogene. 2003, 22 (23): 3645-3654. 10.1038/sj.onc.1206477.

    CAS  PubMed  Google Scholar 

  12. Miyashita T, Reed JC: Tumor suppressor p53 is a direct transcriptional activator of the human bax gene. Cell. 1995, 80 (2): 293-299. 10.1016/0092-8674(95)90412-3.

    CAS  PubMed  Google Scholar 

  13. Nakano K, Vousden KH: PUMA, a novel proapoptotic gene, is induced by p53. Mol Cell. 2001, 7 (3): 683-694. 10.1016/S1097-2765(01)00214-3.

    CAS  PubMed  Google Scholar 

  14. Oda E, Ohki R, Murasawa H, Nemoto J, Shibue T, Yamashita T, Tokino T, Taniguchi T, Tanaka N: Noxa, a BH3-only member of the Bcl-2 family and candidate mediator of p53-induced apoptosis. Science. 2000, 288 (5468): 1053-1058. 10.1126/science.288.5468.1053.

    CAS  PubMed  Google Scholar 

  15. Yu J, Zhang L, Hwang PM, Kinzler KW, Vogelstein B: PUMA induces the rapid apoptosis of colorectal cancer cells. Mol Cell. 2001, 7 (3): 673-682. 10.1016/S1097-2765(01)00213-1.

    CAS  PubMed  Google Scholar 

  16. Chipuk JE, Kuwana T, Bouchier-Hayes L, Droin NM, Newmeyer DD, Schuler M, Green DR: Direct activation of Bax by p53 mediates mitochondrial membrane permeabilization and apoptosis. Science. 2004, 303 (5660): 1010-1014. 10.1126/science.1092734.

    CAS  PubMed  Google Scholar 

  17. Leu JI, Dumont P, Hafey M, Murphy ME, George DL: Mitochondrial p53 activates Bak and causes disruption of a Bak-Mcl1 complex. Nat Cell Biol. 2004, 6 (5): 443-450. 10.1038/ncb1123.

    CAS  PubMed  Google Scholar 

  18. Mihara M, Erster S, Zaika A, Petrenko O, Chittenden T, Pancoska P, Moll UM: p53 has a direct apoptogenic role at the mitochondria. Mol Cell. 2003, 11 (3): 577-590. 10.1016/S1097-2765(03)00050-9.

    CAS  PubMed  Google Scholar 

  19. Mihara M, Moll UM: Detection of mitochondrial localization of p53. Methods Mol Biol. 2003, 234: 203-209.

    CAS  PubMed  Google Scholar 

  20. Kitamura H, Yazawa T, Sato H, Okudela K, Shimoyamada H: Small cell lung cancer: significance of RB alterations and TTF-1 expression in its carcinogenesis, phenotype, and biology. Endocr Pathol. 2009, 20 (2): 101-107. 10.1007/s12022-009-9072-4.

    CAS  PubMed  Google Scholar 

  21. Lee WH, Shew JY, Hong FD, Sery TW, Donoso LA, Young LJ, Bookstein R, Lee EY: The retinoblastoma susceptibility gene encodes a nuclear phosphoprotein associated with DNA binding activity. Nature. 1987, 329 (6140): 642-645. 10.1038/329642a0.

    CAS  PubMed  Google Scholar 

  22. Miller CW, Aslo A, Won A, Tan M, Lampkin B, Koeffler HP: Alterations of the p53, Rb and MDM2 genes in osteosarcoma. J Cancer Res Clin Oncol. 1996, 122 (9): 559-565. 10.1007/BF01213553.

    CAS  PubMed  Google Scholar 

  23. Wang NP, To H, Lee WH, Lee EY: Tumor suppressor activity of RB and p53 genes in human breast carcinoma cells. Oncogene. 1993, 8 (2): 279-288.

    CAS  PubMed  Google Scholar 

  24. Knudsen ES, Wang JY: Targeting the RB-pathway in cancer therapy. Clin Cancer Res. 2010, 16 (4): 1094-1099. 10.1158/1078-0432.CCR-09-0787.

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Harbour JW, Dean DC: The Rb/E2F pathway: expanding roles and emerging paradigms. Genes Dev. 2000, 14 (19): 2393-2409. 10.1101/gad.813200.

    CAS  PubMed  Google Scholar 

  26. Araki K, Kawauchi K, Tanaka N: IKK/NF-kappaB signaling pathway inhibits cell-cycle progression by a novel Rb-independent suppression system for E2F transcription factors. Oncogene. 2008, 27 (43): 5696-5705. 10.1038/onc.2008.184.

    CAS  PubMed  Google Scholar 

  27. Bowen C, Spiegel S, Gelmann EP: Radiation-induced apoptosis mediated by retinoblastoma protein. Cancer Res. 1998, 58 (15): 3275-3281.

    CAS  PubMed  Google Scholar 

  28. Bowen C, Birrer M, Gelmann EP: Retinoblastoma protein-mediated apoptosis after gamma-irradiation. J Biol Chem. 2002, 277 (47): 44969-44979. 10.1074/jbc.M202000200.

    CAS  PubMed  Google Scholar 

  29. Knudsen KE, Weber E, Arden KC, Cavenee WK, Feramisco JR, Knudsen ES: The retinoblastoma tumor suppressor inhibits cellular proliferation through two distinct mechanisms: inhibition of cell cycle progression and induction of cell death. Oncogene. 1999, 18 (37): 5239-5245. 10.1038/sj.onc.1202910.

    CAS  PubMed  Google Scholar 

  30. Almasan A, Yin Y, Kelly RE, Lee EY, Bradley A, Li W, Bertino JR, Wahl GM: Deficiency of retinoblastoma protein leads to inappropriate S-phase entry, activation of E2F-responsive genes, and apoptosis. Proc Natl Acad Sci U S A. 1995, 92 (12): 5436-5440. 10.1073/pnas.92.12.5436.

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Bosco EE, Mayhew CN, Hennigan RF, Sage J, Jacks T, Knudsen ES: RB signaling prevents replication-dependent DNA double-strand breaks following genotoxic insult. Nucleic Acids Res. 2004, 32 (1): 25-34. 10.1093/nar/gkg919.

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Knudsen KE, Booth D, Naderi S, Sever-Chroneos Z, Fribourg AF, Hunton IC, Feramisco JR, Wang JY, Knudsen ES: RB-dependent S-phase response to DNA damage. Mol Cell Biol. 2000, 20 (20): 7751-7763. 10.1128/MCB.20.20.7751-7763.2000.

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Ianari A, Natale T, Calo E, Ferretti E, Alesse E, Screpanti I, Haigis K, Gulino A, Lees JA: Proapoptotic function of the retinoblastoma tumor suppressor protein. Cancer Cell. 2009, 15 (3): 184-194. 10.1016/j.ccr.2009.01.026.

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Ferecatu I, Le Floch N, Bergeaud M, Rodriguez-Enfedaque A, Rincheval V, Oliver L, Vallette FM, Mignotte B, Vayssiere JL: Evidence for a mitochondrial localization of the retinoblastoma protein. BMC Cell Biol. 2009, 10: 50-10.1186/1471-2121-10-50.

    PubMed  PubMed Central  Google Scholar 

  35. Hilgendorf KI, Leshchiner ES, Nedelcu S, Maynard MA, Calo E, Ianari A, Walensky LD, Lees JA: The retinoblastoma protein induces apoptosis directly at the mitochondria. Genes Dev. 2013, 27 (9): 1003-1015. 10.1101/gad.211326.112.

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Macleod KF, Hu Y, Jacks T: Loss of Rb activates both p53-dependent and independent cell death pathways in the developing mouse nervous system. EMBO J. 1996, 15 (22): 6178-6188.

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Morgenbesser SD, Williams BO, Jacks T, DePinho RA: p53-dependent apoptosis produced by Rb-deficiency in the developing mouse lens. Nature. 1994, 371 (6492): 72-74. 10.1038/371072a0.

    CAS  PubMed  Google Scholar 

  38. Gao J, Aksoy BA, Dogrusoz U, Dresdner G, Gross B, Sumer SO, Sun Y, Jacobsen A, Sinha R, Larsson E, Cerami E, Sander C, Schultz N: Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal. 2013, 6 (269): l1-

    Google Scholar 

  39. Cerami E, Gao J, Dogrusoz U, Gross BE, Sumer SO, Aksoy BA, Jacobsen A, Byrne CJ, Heuer ML, Larsson E, Antipin Y, Reva B, Goldberg AP, Sander C, Schultz N: The cBio cancer genomics portal: an open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012, 2 (5): 401-404. 10.1158/2159-8290.CD-12-0095.

    PubMed  Google Scholar 

  40. Leslie NR, Brunton VG: Cell biology. Where is PTEN?. Science. 2013, 341 (6144): 355-356. 10.1126/science.1242541.

    CAS  PubMed  Google Scholar 

  41. Hopkins BD, Hodakoski C, Barrows D, Mense SM, Parsons RE: PTEN function: the long and the short of it. Trends Biochem Sci. 2014, 39 (4): 183-190. 10.1016/j.tibs.2014.02.006.

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Stambolic V, Suzuki A, de la Pompa JL, Brothers GM, Mirtsos C, Sasaki T, Ruland J, Penninger JM, Siderovski DP, Mak TW: Negative regulation of PKB/Akt-dependent cell survival by the tumor suppressor PTEN. Cell. 1998, 95 (1): 29-39. 10.1016/S0092-8674(00)81780-8.

    CAS  PubMed  Google Scholar 

  43. Datta SR, Brunet A, Greenberg ME: Cellular survival: a play in three Akts. Genes Dev. 1999, 13 (22): 2905-2927. 10.1101/gad.13.22.2905.

    CAS  PubMed  Google Scholar 

  44. Dijkers PF, Medema RH, Lammers JW, Koenderman L, Coffer PJ: Expression of the pro-apoptotic Bcl-2 family member Bim is regulated by the forkhead transcription factor FKHR-L1. Curr Biol. 2000, 10 (19): 1201-1204. 10.1016/S0960-9822(00)00728-4.

    CAS  PubMed  Google Scholar 

  45. Dudgeon C, Wang P, Sun X, Peng R, Sun Q, Yu J, Zhang L: PUMA induction by FoxO3a mediates the anticancer activities of the broad-range kinase inhibitor UCN-01. Mol Cancer Ther. 2010, 9 (11): 2893-2902. 10.1158/1535-7163.MCT-10-0635.

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Dan HC, Sun M, Kaneko S, Feldman RI, Nicosia SV, Wang HG, Tsang BK, Cheng JQ: Akt phosphorylation and stabilization of X-linked inhibitor of apoptosis protein (XIAP). J Biol Chem. 2004, 279 (7): 5405-5412. 10.1074/jbc.M312044200.

    CAS  PubMed  Google Scholar 

  47. Datta SR, Dudek H, Tao X, Masters S, Fu H, Gotoh Y, Greenberg ME: Akt phosphorylation of BAD couples survival signals to the cell-intrinsic death machinery. Cell. 1997, 91 (2): 231-241. 10.1016/S0092-8674(00)80405-5.

    CAS  PubMed  Google Scholar 

  48. del Peso L, Gonzalez-Garcia M, Page C, Herrera R, Nunez G: Interleukin-3-induced phosphorylation of BAD through the protein kinase Akt. Science. 1997, 278 (5338): 687-689. 10.1126/science.278.5338.687.

    CAS  PubMed  Google Scholar 

  49. Capon DJ, Seeburg PH, McGrath JP, Hayflick JS, Edman U, Levinson AD, Goeddel DV: Activation of Ki-ras2 gene in human colon and lung carcinomas by two different point mutations. Nature. 1983, 304 (5926): 507-513. 10.1038/304507a0.

    CAS  PubMed  Google Scholar 

  50. Chang EH, Furth ME, Scolnick EM, Lowy DR: Tumorigenic transformation of mammalian cells induced by a normal human gene homologous to the oncogene of Harvey murine sarcoma virus. Nature. 1982, 297 (5866): 479-483. 10.1038/297479a0.

    CAS  PubMed  Google Scholar 

  51. Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S, Teague J, Woffendin H, Garnett MJ, Bottomley W, Davis N, Dicks E, Ewing R, Floyd Y, Gray K, Hall S, Hawes R, Hughes J, Kosmidou V, Menzies A, Mould C, Parker A, Stevens C, Watt S, Hooper S, Wilson R, Jayatilake H, Gusterson BA, Cooper C, Shipley J: Mutations of the BRAF gene in human cancer. Nature. 2002, 417 (6892): 949-954. 10.1038/nature00766.

    CAS  PubMed  Google Scholar 

  52. Downward J: Targeting RAS signalling pathways in cancer therapy. Nat Rev Cancer. 2003, 3 (1): 11-22. 10.1038/nrc969.

    CAS  PubMed  Google Scholar 

  53. Pratilas CA, Solit DB: Targeting the mitogen-activated protein kinase pathway: physiological feedback and drug response. Clin Cancer Res. 2010, 16 (13): 3329-3334. 10.1158/1078-0432.CCR-09-3064.

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Kinoshita T, Yokota T, Arai K, Miyajima A: Regulation of Bcl-2 expression by oncogenic Ras protein in hematopoietic cells. Oncogene. 1995, 10 (11): 2207-2212.

    CAS  PubMed  Google Scholar 

  55. Domina AM, Vrana JA, Gregory MA, Hann SR, Craig RW: MCL1 is phosphorylated in the PEST region and stabilized upon ERK activation in viable cells, and at additional sites with cytotoxic okadaic acid or taxol. Oncogene. 2004, 23 (31): 5301-5315. 10.1038/sj.onc.1207692.

    CAS  PubMed  Google Scholar 

  56. Biswas SC, Greene LA: Nerve growth factor (NGF) down-regulates the Bcl-2 homology 3 (BH3) domain-only protein Bim and suppresses its proapoptotic activity by phosphorylation. J Biol Chem. 2002, 277 (51): 49511-49516. 10.1074/jbc.M208086200.

    CAS  PubMed  Google Scholar 

  57. Goldstein NB, Johannes WU, Gadeliya AV, Green MR, Fujita M, Norris DA, Shellman YG: Active N-Ras and B-Raf inhibit anoikis by downregulating Bim expression in melanocytic cells. J Invest Dermatol. 2009, 129 (2): 432-437. 10.1038/jid.2008.227.

    CAS  PubMed  Google Scholar 

  58. Hubner A, Barrett T, Flavell RA, Davis RJ: Multisite phosphorylation regulates Bim stability and apoptotic activity. Mol Cell. 2008, 30 (4): 415-425. 10.1016/j.molcel.2008.03.025.

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Jiang CC, Lai F, Tay KH, Croft A, Rizos H, Becker TM, Yang F, Liu H, Thorne RF, Hersey P, Zhang XD: Apoptosis of human melanoma cells induced by inhibition of B-RAFV600E involves preferential splicing of bimS. Cell Death Dis. 2010, 1: e69-10.1038/cddis.2010.48.

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Paraiso KH, Xiang Y, Rebecca VW, Abel EV, Chen YA, Munko AC, Wood E, Fedorenko IV, Sondak VK, Anderson AR, Ribas A, Palma MD, Nathanson KL, Koomen JM, Messina JL, Smalley KS: PTEN loss confers BRAF inhibitor resistance to melanoma cells through the suppression of BIM expression. Cancer Res. 2011, 71 (7): 2750-2760. 10.1158/0008-5472.CAN-10-2954.

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Serasinghe MN, Missert DJ, Asciolla JJ, Podgrabinska S, Wieder SY, Izadmehr S, Belbin G, Skobe M, Chipuk JE: Anti-apoptotic BCL-2 proteins govern cellular outcome following B-RAF inhibition and can be targeted to reduce resistance. Oncogene. 2014, 0: doi:10.1038/onc.2014.21

    Google Scholar 

  62. Joseph EW, Pratilas CA, Poulikakos PI, Tadi M, Wang W, Taylor BS, Halilovic E, Persaud Y, Xing F, Viale A, Tsai J, Chapman PB, Bollag G, Solit DB, Rosen N: The RAF inhibitor PLX4032 inhibits ERK signaling and tumor cell proliferation in a V600E BRAF-selective manner. Proc Natl Acad Sci U S A. 2010, 107 (33): 14903-14908. 10.1073/pnas.1008990107.

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Allan LA, Morrice N, Brady S, Magee G, Pathak S, Clarke PR: Inhibition of caspase-9 through phosphorylation at Thr 125 by ERK MAPK. Nat Cell Biol. 2003, 5 (7): 647-654. 10.1038/ncb1005.

    CAS  PubMed  Google Scholar 

  64. Fang X, Yu S, Eder A, Mao M, Bast RC, Boyd D, Mills GB: Regulation of BAD phosphorylation at serine 112 by the Ras-mitogen-activated protein kinase pathway. Oncogene. 1999, 18 (48): 6635-6640. 10.1038/sj.onc.1203076.

    CAS  PubMed  Google Scholar 

  65. Harada H, Andersen JS, Mann M, Terada N, Korsmeyer SJ: p70S6 kinase signals cell survival as well as growth, inactivating the pro-apoptotic molecule BAD. Proc Natl Acad Sci U S A. 2001, 98 (17): 9666-9670. 10.1073/pnas.171301998.

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Kim J, Parrish AB, Kurokawa M, Matsuura K, Freel CD, Andersen JL, Johnson CE, Kornbluth S: Rsk-mediated phosphorylation and 14-3-3varepsilon binding of Apaf-1 suppresses cytochrome c-induced apoptosis. EMBO J. 2012, 31 (5): 1279-1292. 10.1038/emboj.2011.491.

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Nilsson JA, Cleveland JL: Myc pathways provoking cell suicide and cancer. Oncogene. 2003, 22 (56): 9007-9021. 10.1038/sj.onc.1207261.

    CAS  PubMed  Google Scholar 

  68. Green DR, Evan GI: A matter of life and death. Cancer Cell. 2002, 1 (1): 19-30. 10.1016/S1535-6108(02)00024-7.

    CAS  PubMed  Google Scholar 

  69. Hemann MT, Bric A, Teruya-Feldstein J, Herbst A, Nilsson JA, Cordon-Cardo C, Cleveland JL, Tansey WP, Lowe SW: Evasion of the p53 tumour surveillance network by tumour-derived MYC mutants. Nature. 2005, 436 (7052): 807-811. 10.1038/nature03845.

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Terunuma A, Putluri N, Mishra P, Mathe EA, Dorsey TH, Yi M, Wallace TA, Issaq HJ, Zhou M, Killian JK, Stevenson HS, Karoly ED, Chan K, Samanta S, Prieto D, Hsu TY, Kurley SJ, Putluri V, Sonavane R, Edelman DC, Wulff J, Starks AM, Yang Y, Kittles RA, Yfantis HG, Lee DH, Ioffe OB, Schiff R, Stephens RM, Meltzer PS, Veenstra TD, Westbrook TF, Sreekumar A, Ambs S: MYC-driven accumulation of 2-hydroxyglutarate is associated with breast cancer prognosis. J Clin Invest. 2014, 124 (1): 398-412. 10.1172/JCI71180.

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Tsujimoto Y, Gorham J, Cossman J, Jaffe E, Croce CM: The T(14,18) chromosome translocations involved in B-cell neoplasms result from mistakes in VDJ joining. Science. 1985, 229 (4720): 1390-1392. 10.1126/science.3929382.

    CAS  PubMed  Google Scholar 

  72. Vaux DL: Immunopathology of apoptosis—introduction and overview. Springer Semin Immunopathol. 1998, 19 (3): 271-278. 10.1007/BF00787224.

    CAS  PubMed  Google Scholar 

  73. Campos L, Rouault JP, Sabido O, Oriol P, Roubi N, Vasselon C, Archimbaud E, Magaud JP, Guyotat D: High expression of bcl-2 protein in acute myeloid leukemia cells is associated with poor response to chemotherapy. Blood. 1993, 81 (11): 3091-3096.

    CAS  PubMed  Google Scholar 

  74. Frommel TO, Yong S, Zarling EJ: Immunohistochemical evaluation of Bcl-2 gene family expression in liver of hepatitis C and cirrhotic patients: a novel mechanism to explain the high incidence of hepatocarcinoma in cirrhotics. Am J Gastroenterol. 1999, 94 (1): 178-182. 10.1111/j.1572-0241.1999.00792.x.

    CAS  PubMed  Google Scholar 

  75. Lai C, Grant C, Dunleavy K: Interpreting MYC and BCL2 in diffuse large B-cell lymphoma. Leuk Lymphoma. 2013, 54 (10): 2091-2092. 10.3109/10428194.2013.806803.

    CAS  PubMed  Google Scholar 

  76. Steinert DM, Oyarzo M, Wang X, Choi H, Thall PF, Medeiros LJ, Raymond AK, Benjamin RS, Zhang W, Trent JC: Expression of Bcl-2 in gastrointestinal stromal tumors: correlation with progression-free survival in 81 patients treated with imatinib mesylate. Cancer. 2006, 106 (7): 1617-1623. 10.1002/cncr.21781.

    PubMed  Google Scholar 

  77. Renault TT, Chipuk JE: Death upon a kiss: mitochondrial outer membrane composition and organelle communication govern sensitivity to BAK/BAX-dependent apoptosis. Chem Biol. 2014, 21 (1): 114-123. 10.1016/j.chembiol.2013.10.009.

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Kuwana T, Mackey MR, Perkins G, Ellisman MH, Latterich M, Schneiter R, Green DR, Newmeyer DD: Bid, Bax, and lipids cooperate to form supramolecular openings in the outer mitochondrial membrane. Cell. 2002, 111 (3): 331-342. 10.1016/S0092-8674(02)01036-X.

    CAS  PubMed  Google Scholar 

  79. Chipuk JE, McStay GP, Bharti A, Kuwana T, Clarke CJ, Siskind LJ, Obeid LM, Green DR: Sphingolipid metabolism cooperates with BAK and BAX to promote the mitochondrial pathway of apoptosis. Cell. 2012, 148 (5): 988-1000. 10.1016/j.cell.2012.01.038.

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Kuwana T, Bouchier-Hayes L, Chipuk JE, Bonzon C, Sullivan BA, Green DR, Newmeyer DD: BH3 domains of BH3-only proteins differentially regulate Bax-mediated mitochondrial membrane permeabilization both directly and indirectly. Mol Cell. 2005, 17 (4): 525-535. 10.1016/j.molcel.2005.02.003.

    CAS  PubMed  Google Scholar 

  81. 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 (3): 183-192. 10.1016/S1535-6108(02)00127-7.

    CAS  PubMed  Google Scholar 

  82. Gavathiotis E, Suzuki M, Davis ML, Pitter K, Bird GH, Katz SG, Tu HC, Kim H, Cheng EH, Tjandra N, Walensky LD: BAX activation is initiated at a novel interaction site. Nature. 2008, 455 (7216): 1076-1081. 10.1038/nature07396.

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Desagher S, Osen-Sand A, Nichols A, Eskes R, Montessuit S, Lauper S, Maundrell K, Antonsson B, Martinou JC: Bid-induced conformational change of Bax is responsible for mitochondrial cytochrome c release during apoptosis. J Cell Biol. 1999, 144 (5): 891-901. 10.1083/jcb.144.5.891.

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Czabotar PE, Westphal D, Dewson G, Ma S, Hockings C, Fairlie WD, Lee EF, Yao S, Robin AY, Smith BJ, Huang DC, Kluck RM, Adams JM, Colman PM: Bax crystal structures reveal how BH3 domains activate Bax and nucleate its oligomerization to induce apoptosis. Cell. 2013, 152 (3): 519-531. 10.1016/j.cell.2012.12.031.

    CAS  PubMed  Google Scholar 

  85. Chipuk JE, Fisher JC, Dillon CP, Kriwacki RW, Kuwana T, Green DR: Mechanism of apoptosis induction by inhibition of the anti-apoptotic BCL-2 proteins. Proc Natl Acad Sci U S A. 2008, 105 (51): 20327-20332. 10.1073/pnas.0808036105.

    CAS  PubMed  PubMed Central  Google Scholar 

  86. Renault TT, Chipuk JE: Getting away with murder: how does the BCL-2 family of proteins kill with immunity?. Ann N Y Acad Sci. 2013, 1285: 59-79. 10.1111/nyas.12045.

    CAS  PubMed  PubMed Central  Google Scholar 

  87. Hanada M, Delia D, Aiello A, Stadtmauer E, Reed JC: bcl-2 gene hypomethylation and high-level expression in B-cell chronic lymphocytic leukemia. Blood. 1993, 82 (6): 1820-1828.

    CAS  PubMed  Google Scholar 

  88. Certo M, Del Gaizo MV, Nishino M, Wei G, Korsmeyer S, Armstrong SA, Letai A: Mitochondria primed by death signals determine cellular addiction to antiapoptotic BCL-2 family members. Cancer Cell. 2006, 9 (5): 351-365. 10.1016/j.ccr.2006.03.027.

    CAS  PubMed  Google Scholar 

  89. Ni Chonghaile T, Sarosiek KA, Vo TT, Ryan JA, Tammareddi A, Moore Vdel G, Deng J, Anderson KC, Richardson P, Tai YT, Mitsiades CS, Matulonis UA, Drapkin R, Stone R, Deangelo DJ, McConkey DJ, Sallan SE, Silverman L, Hirsch MS, Carrasco DR, Letai A: Pretreatment mitochondrial priming correlates with clinical response to cytotoxic chemotherapy. Science. 2011, 334 (6059): 1129-1133. 10.1126/science.1206727.

    PubMed  Google Scholar 

  90. Ryan JA, Brunelle JK, Letai A: Heightened mitochondrial priming is the basis for apoptotic hypersensitivity of CD4+ CD8+ thymocytes. Proc Natl Acad Sci U S A. 2010, 107 (29): 12895-12900. 10.1073/pnas.0914878107.

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Cimmino A, Calin GA, Fabbri M, Iorio MV, Ferracin M, Shimizu M, Wojcik SE, Aqeilan RI, Zupo S, Dono M, Rassenti L, Alder H, Volinia S, Liu CG, Kipps TJ, Negrini M, Croce CM: miR-15 and miR-16 induce apoptosis by targeting BCL2. Proc Natl Acad Sci U S A. 2005, 102 (39): 13944-13949. 10.1073/pnas.0506654102.

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Zhu DX, Zhu W, Fang C, Fan L, Zou ZJ, Wang YH, Liu P, Hong M, Miao KR, Liu P, Xu W, Li JY: miR-181a/b significantly enhances drug sensitivity in chronic lymphocytic leukemia cells via targeting multiple anti-apoptosis genes. Carcinogenesis. 2012, 33 (7): 1294-1301. 10.1093/carcin/bgs179.

    CAS  PubMed  Google Scholar 

  93. She QB, Solit DB, Ye Q, O'Reilly KE, Lobo J, Rosen N: The BAD protein integrates survival signaling by EGFR/MAPK and PI3K/Akt kinase pathways in PTEN-deficient tumor cells. Cancer Cell. 2005, 8 (4): 287-297. 10.1016/j.ccr.2005.09.006.

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Paterson A, Mockridge CI, Adams JE, Krysov S, Potter KN, Duncombe AS, Cook SJ, Stevenson FK, Packham G: Mechanisms and clinical significance of BIM phosphorylation in chronic lymphocytic leukemia. Blood. 2012, 119 (7): 1726-1736. 10.1182/blood-2011-07-367417.

    CAS  PubMed  Google Scholar 

  95. Kitada S, Leone M, Sareth S, Zhai D, Reed JC, Pellecchia M: Discovery, characterization, and structure-activity relationships studies of proapoptotic polyphenols targeting B-cell lymphocyte/leukemia-2 proteins. J Med Chem. 2003, 46 (20): 4259-4264. 10.1021/jm030190z.

    CAS  PubMed  Google Scholar 

  96. Mohammad RM, Goustin AS, Aboukameel A, Chen B, Banerjee S, Wang G, Nikolovska-Coleska Z, Wang S, Al-Katib A: Preclinical studies of TW-37, a new nonpeptidic small-molecule inhibitor of Bcl-2, in diffuse large cell lymphoma xenograft model reveal drug action on both Bcl-2 and Mcl-1. Clin Cancer Res. 2007, 13 (7): 2226-2235. 10.1158/1078-0432.CCR-06-1574.

    CAS  PubMed  Google Scholar 

  97. Wang G, Nikolovska-Coleska Z, Yang CY, Wang R, Tang G, Guo J, Shangary S, Qiu S, Gao W, Yang D, Meagher J, Stuckey J, Krajewski K, Jiang S, Roller PP, Abaan HO, Tomita Y, Wang S: Structure-based design of potent small-molecule inhibitors of anti-apoptotic Bcl-2 proteins. J Med Chem. 2006, 49 (21): 6139-6142. 10.1021/jm060460o.

    CAS  PubMed  Google Scholar 

  98. Wei J, Kitada S, Stebbins JL, Placzek W, Zhai D, Wu B, Rega MF, Zhang Z, Cellitti J, Yang L, Dahl R, Reed JC, Pellecchia M: Synthesis and biological evaluation of Apogossypolone derivatives as pan-active inhibitors of antiapoptotic B-cell lymphoma/leukemia-2 (Bcl-2) family proteins. J Med Chem. 2010, 53 (22): 8000-8011. 10.1021/jm100746q.

    CAS  PubMed  PubMed Central  Google Scholar 

  99. Oltersdorf T, Elmore SW, Shoemaker AR, Armstrong RC, Augeri DJ, Belli BA, Bruncko M, Deckwerth TL, Dinges J, Hajduk PJ, Joseph MK, Kitada S, Korsmeyer SJ, Kunzer AR, Letai A, Li C, Mitten MJ, Nettesheim DG, Ng S, Nimmer PM, O'Connor JM, Oleksijew A, Petros AM, Reed JC, Shen W, Tahir SK, Thompson CB, Tomaselli KJ, Wang B, Wendt MD, Zhang H, Fesik SW, Rosenberg SH: An inhibitor of Bcl-2 family proteins induces regression of solid tumours. Nature. 2005, 435 (7042): 677-681. 10.1038/nature03579.

    CAS  PubMed  Google Scholar 

  100. Mason EF, Rathmell JC: Cell metabolism: an essential link between cell growth and apoptosis. Biochim Biophys Acta. 2011, 1813 (4): 645-654. 10.1016/j.bbamcr.2010.08.011.

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Zhang H, Nimmer PM, Tahir SK, Chen J, Fryer RM, Hahn KR, Iciek LA, Morgan SJ, Nasarre MC, Nelson R, Preusser LC, Reinhart GA, Smith ML, Rosenberg SH, Elmore SW, Tse C: Bcl-2 family proteins are essential for platelet survival. Cell Death Differ. 2007, 14 (5): 943-951.

    CAS  PubMed  Google Scholar 

  102. Souers AJ, Leverson JD, Boghaert ER, Ackler SL, Catron ND, Chen J, Dayton BD, Ding H, Enschede SH, Fairbrother WJ, Huang DC, Hymowitz SG, Jin S, Khaw SL, Kovar PJ, Lam LT, Lee J, Maecker HL, Marsh KC, Mason KD, Mitten MJ, Nimmer PM, Oleksijew A, Park CH, Park CM, Phillips DC, Roberts AW, Sampath D, Seymour JF, Smith ML: ABT-199, a potent and selective BCL-2 inhibitor, achieves antitumor activity while sparing platelets. Nat Med. 2013, 19 (2): 202-208. 10.1038/nm.3048.

    CAS  PubMed  Google Scholar 

  103. Vandenberg CJ, Cory S: ABT-199, a new Bcl-2-specific BH3 mimetic, has in vivo efficacy against aggressive Myc-driven mouse lymphomas without provoking thrombocytopenia. Blood. 2013, 121 (12): 2285-2288. 10.1182/blood-2013-01-475855.

    CAS  PubMed  PubMed Central  Google Scholar 

  104. Cohen NA, Stewart ML, Gavathiotis E, Tepper JL, Bruekner SR, Koss B, Opferman JT, Walensky LD: A competitive stapled peptide screen identifies a selective small molecule that overcomes MCL-1-dependent leukemia cell survival. Chem Biol. 2012, 19 (9): 1175-1186. 10.1016/j.chembiol.2012.07.018.

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Gautier F, Guillemin Y, Cartron PF, Gallenne T, Cauquil N, Le Diguarher T, Casara P, Vallette FM, Manon S, Hickman JA, Geneste O, Juin P: Bax activation by engagement with, then release from, the BH3 binding site of Bcl-xL. Mol Cell Biol. 2011, 31 (4): 832-844. 10.1128/MCB.00161-10.

    CAS  PubMed  PubMed Central  Google Scholar 

  106. Lessene G, Czabotar PE, Sleebs BE, Zobel K, Lowes KN, Adams JM, Baell JB, Colman PM, Deshayes K, Fairbrother WJ, Flygare JA, Gibbons P, Kersten WJ, Kulasegaram S, Moss RM, Parisot JP, Smith BJ, Street IP, Yang H, Huang DC, Watson KG: Structure-guided design of a selective BCL-X(L) inhibitor. Nat Chem Biol. 2013, 9 (6): 390-397. 10.1038/nchembio.1246.

    CAS  PubMed  Google Scholar 

  107. Deng J, Carlson N, Takeyama K, Dal Cin P, Shipp M, Letai A: BH3 profiling identifies three distinct classes of apoptotic blocks to predict response to ABT-737 and conventional chemotherapeutic agents. Cancer Cell. 2007, 12 (2): 171-185. 10.1016/j.ccr.2007.07.001.

    CAS  PubMed  Google Scholar 

  108. Ryan J, Letai A: BH3 profiling in whole cells by fluorimeter or FACS. Methods. 2013, 61 (2): 156-164. 10.1016/j.ymeth.2013.04.006.

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Renault TT, Elkholi R, Bharti A, Chipuk JE: BH3 mimetics demonstrate differential activities dependent upon the functional repertoire of pro- and anti-apoptotic BCL-2 family proteins. J Biol Chem. 2014

    Google Scholar 

  110. Gavathiotis E, Reyna DE, Bellairs JA, Leshchiner ES, Walensky LD: Direct and selective small-molecule activation of proapoptotic BAX. Nat Chem Biol. 2012, 8 (7): 639-645. 10.1038/nchembio.995.

    CAS  PubMed  PubMed Central  Google Scholar 

  111. Del Gaizo MV, Brown JR, Certo M, Love TM, Novina CD, Letai A: Chronic lymphocytic leukemia requires BCL2 to sequester prodeath BIM, explaining sensitivity to BCL2 antagonist ABT-737. J Clin Invest. 2007, 117 (1): 112-121. 10.1172/JCI28281.

    Google Scholar 

  112. Bean GR, Ganesan YT, Dong Y, Takeda S, Liu H, Chan PM, Huang Y, Chodosh LA, Zambetti GP, Hsieh JJ, Cheng EH: PUMA and BIM are required for oncogene inactivation-induced apoptosis. Sci Signal. 2013, 6 (268): ra20-

    PubMed  PubMed Central  Google Scholar 

  113. Anvekar RA, Asciolla JJ, Missert DJ, Chipuk JE: Born to be alive: a role for the BCL-2 family in melanoma tumor cell survival, apoptosis, and treatment. Front Oncol. 2011, 1 (34):

  114. Tait SW, Green DR: Mitochondria and cell death: outer membrane permeabilization and beyond. Nat Rev Mol Cell Biol. 2010, 11 (9): 621-632. 10.1038/nrm2952.

    CAS  PubMed  Google Scholar 

  115. Frezza C, Cipolat S, Martins de Brito O, Micaroni M, Beznoussenko GV, Rudka T, Bartoli D, Polishuck RS, Danial NN, De Strooper B, Scorrano L: OPA1 controls apoptotic cristae remodeling independently from mitochondrial fusion. Cell. 2006, 126 (1): 177-189. 10.1016/j.cell.2006.06.025.

    CAS  PubMed  Google Scholar 

  116. Yamaguchi R, Lartigue L, Perkins G, Scott RT, Dixit A, Kushnareva Y, Kuwana T, Ellisman MH, Newmeyer DD: Opa1-mediated cristae opening is Bax/Bak and BH3 dependent, required for apoptosis, and independent of Bak oligomerization. Mol Cell. 2008, 31 (4): 557-569. 10.1016/j.molcel.2008.07.010.

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Scorrano L, Ashiya M, Buttle K, Weiler S, Oakes SA, Mannella CA, Korsmeyer SJ: A distinct pathway remodels mitochondrial cristae and mobilizes cytochrome c during apoptosis. Dev Cell. 2002, 2 (1): 55-67. 10.1016/S1534-5807(01)00116-2.

    CAS  PubMed  Google Scholar 

  118. Segawa K, Kurata S, Yanagihashi Y, Brummelkamp TR, Matsuda F, Nagata S: Caspase-mediated cleavage of phospholipid flippase for apoptotic phosphatidylserine exposure. Science. 2014, 344 (6188): 1164-1168. 10.1126/science.1252809.

    CAS  PubMed  Google Scholar 

  119. Poon IK, Lucas CD, Rossi AG, Ravichandran KS: Apoptotic cell clearance: basic biology and therapeutic potential. Nat Rev Immunol. 2014, 14 (3): 166-180. 10.1038/nri3607.

    CAS  PubMed  PubMed Central  Google Scholar 

  120. Vempati UD, Han X, Moraes CT: Lack of cytochrome c in mouse fibroblasts disrupts assembly/stability of respiratory complexes I and IV. J Biol Chem. 2009, 284 (7): 4383-4391. 10.1074/jbc.M805972200.

    CAS  PubMed  PubMed Central  Google Scholar 

  121. Hao Z, Duncan GS, Chang CC, Elia A, Fang M, Wakeham A, Okada H, Calzascia T, Jang Y, You-Ten A, Yeh WC, Ohashi P, Wang X, Mak TW: Specific ablation of the apoptotic functions of cytochrome C reveals a differential requirement for cytochrome C and Apaf-1 in apoptosis. Cell. 2005, 121 (4): 579-591. 10.1016/j.cell.2005.03.016.

    CAS  PubMed  Google Scholar 

  122. Li P, Nijhawan D, Budihardjo I, Srinivasula SM, Ahmad M, Alnemri ES, Wang X: Cytochrome c and dATP-dependent formation of Apaf-1/caspase-9 complex initiates an apoptotic protease cascade. Cell. 1997, 91 (4): 479-489. 10.1016/S0092-8674(00)80434-1.

    CAS  PubMed  Google Scholar 

  123. Hakem R, Hakem A, Duncan GS, Henderson JT, Woo M, Soengas MS, Elia A, de la Pompa JL, Kagi D, Khoo W, Potter J, Yoshida R, Kaufman SA, Lowe SW, Penninger JM, Mak TW: Differential requirement for caspase 9 in apoptotic pathways in vivo. Cell. 1998, 94 (3): 339-352. 10.1016/S0092-8674(00)81477-4.

    CAS  PubMed  Google Scholar 

  124. Honarpour N, Du C, Richardson JA, Hammer RE, Wang X, Herz J: Adult Apaf-1-deficient mice exhibit male infertility. Dev Biol. 2000, 218 (2): 248-258. 10.1006/dbio.1999.9585.

    CAS  PubMed  Google Scholar 

  125. Scott CL, Schuler M, Marsden VS, Egle A, Pellegrini M, Nesic D, Gerondakis S, Nutt SL, Green DR, Strasser A: Apaf-1 and caspase-9 do not act as tumor suppressors in myc-induced lymphomagenesis or mouse embryo fibroblast transformation. J Cell Biol. 2004, 164 (1): 89-96. 10.1083/jcb.200310041.

    CAS  PubMed  PubMed Central  Google Scholar 

  126. Soengas MS, Alarcon RM, Yoshida H, Giaccia AJ, Hakem R, Mak TW, Lowe SW: Apaf-1 and caspase-9 in p53-dependent apoptosis and tumor inhibition. Science. 1999, 284 (5411): 156-159. 10.1126/science.284.5411.156.

    CAS  PubMed  Google Scholar 

  127. Egle A, Harris AW, Bouillet P, Cory S: Bim is a suppressor of Myc-induced mouse B cell leukemia. Proc Natl Acad Sci U S A. 2004, 101 (16): 6164-6169. 10.1073/pnas.0401471101.

    CAS  PubMed  PubMed Central  Google Scholar 

  128. Frenzel A, Labi V, Chmelewskij W, Ploner C, Geley S, Fiegl H, Tzankov A, Villunger A: Suppression of B-cell lymphomagenesis by the BH3-only proteins Bmf and Bad. Blood. 2010, 115 (5): 995-1005. 10.1182/blood-2009-03-212670.

    CAS  PubMed  PubMed Central  Google Scholar 

  129. Salvesen GS, Abrams JM: Caspase activation—stepping on the gas or releasing the brakes? Lessons from humans and flies. Oncogene. 2004, 23 (16): 2774-2784. 10.1038/sj.onc.1207522.

    CAS  PubMed  Google Scholar 

  130. Eckelman BP, Salvesen GS, Scott FL: Human inhibitor of apoptosis proteins: why XIAP is the black sheep of the family. EMBO Rep. 2006, 7 (10): 988-994. 10.1038/sj.embor.7400795.

    CAS  PubMed  PubMed Central  Google Scholar 

  131. Suzuki Y, Imai Y, Nakayama H, Takahashi K, Takio K, Takahashi R: A serine protease, HtrA2, is released from the mitochondria and interacts with XIAP, inducing cell death. Mol Cell. 2001, 8 (3): 613-621. 10.1016/S1097-2765(01)00341-0.

    CAS  PubMed  Google Scholar 

  132. Verhagen AM, Ekert PG, Pakusch M, Silke J, Connolly LM, Reid GE, Moritz RL, Simpson RJ, Vaux DL: Identification of DIABLO, a mammalian protein that promotes apoptosis by binding to and antagonizing IAP proteins. Cell. 2000, 102 (1): 43-53. 10.1016/S0092-8674(00)00009-X.

    CAS  PubMed  Google Scholar 

  133. Okada H, Suh WK, Jin J, Woo M, Du C, Elia A, Duncan GS, Wakeham A, Itie A, Lowe SW, Wang X, Mak TW: Generation and characterization of Smac/DIABLO-deficient mice. Mol Cell Biol. 2002, 22 (10): 3509-3517. 10.1128/MCB.22.10.3509-3517.2002.

    CAS  PubMed  PubMed Central  Google Scholar 

  134. Kempkensteffen C, Hinz S, Christoph F, Krause H, Magheli A, Schrader M, Schostak M, Miller K, Weikert S: Expression levels of the mitochondrial IAP antagonists Smac/DIABLO and Omi/HtrA2 in clear-cell renal cell carcinomas and their prognostic value. J Cancer Res Clin Oncol. 2008, 134 (5): 543-550. 10.1007/s00432-007-0317-7.

    CAS  PubMed  Google Scholar 

  135. Mizutani Y, Nakanishi H, Yamamoto K, Li YN, Matsubara H, Mikami K, Okihara K, Kawauchi A, Bonavida B, Miki T: Downregulation of Smac/DIABLO expression in renal cell carcinoma and its prognostic significance. J Clin Oncol. 2005, 23 (3): 448-454.

    CAS  PubMed  Google Scholar 

  136. Sekimura A, Konishi A, Mizuno K, Kobayashi Y, Sasaki H, Yano M, Fukai I, Fujii Y: Expression of Smac/DIABLO is a novel prognostic marker in lung cancer. Oncol Rep. 2004, 11 (4): 797-802.

    CAS  PubMed  Google Scholar 

  137. Hofmann HS, Simm A, Hammer A, Silber RE, Bartling B: Expression of inhibitors of apoptosis (IAP) proteins in non-small cell human lung cancer. J Cancer Res Clin Oncol. 2002, 128 (10): 554-560. 10.1007/s00432-002-0364-z.

    CAS  PubMed  Google Scholar 

  138. Espinosa M, Cantu D, Lopez CM, De la Garza JG, Maldonado VA, Melendez-Zajgla J: SMAC is expressed de novo in a subset of cervical cancer tumors. BMC Cancer. 2004, 4: 84-10.1186/1471-2407-4-84.

    PubMed  PubMed Central  Google Scholar 

  139. Khodjakov A, Rieder C, Mannella CA, Kinnally KW: Laser micro-irradiation of mitochondria: is there an amplified mitochondrial death signal in neural cells?. Mitochondrion. 2004, 3 (4): 217-227. 10.1016/j.mito.2003.10.002.

    CAS  PubMed  Google Scholar 

  140. Harlin H, Reffey SB, Duckett CS, Lindsten T, Thompson CB: Characterization of XIAP-deficient mice. Mol Cell Biol. 2001, 21 (10): 3604-3608. 10.1128/MCB.21.10.3604-3608.2001.

    CAS  PubMed  PubMed Central  Google Scholar 

  141. Lamkanfi M, Declercq W, Vanden Berghe T, Vandenabeele P: Caspases leave the beaten track: caspase-mediated activation of NF-kappaB. J Cell Biol. 2006, 173 (2): 165-171. 10.1083/jcb.200509092.

    CAS  PubMed  PubMed Central  Google Scholar 

  142. Woo M, Hakem R, Furlonger C, Hakem A, Duncan GS, Sasaki T, Bouchard D, Lu L, Wu GE, Paige CJ, Mak TW: Caspase-3 regulates cell cycle in B cells: a consequence of substrate specificity. Nat Immunol. 2003, 4 (10): 1016-1022. 10.1038/ni976.

    CAS  PubMed  Google Scholar 

  143. Zandy AJ, Lakhani S, Zheng T, Flavell RA, Bassnett S: Role of the executioner caspases during lens development. J Biol Chem. 2005, 280 (34): 30263-30272. 10.1074/jbc.M504007200.

    CAS  PubMed  Google Scholar 

  144. Zermati Y, Garrido C, Amsellem S, Fishelson S, Bouscary D, Valensi F, Varet B, Solary E, Hermine O: Caspase activation is required for terminal erythroid differentiation. J Exp Med. 2001, 193 (2): 247-254. 10.1084/jem.193.2.247.

    CAS  PubMed  PubMed Central  Google Scholar 

  145. Fernando P, Kelly JF, Balazsi K, Slack RS, Megeney LA: Caspase 3 activity is required for skeletal muscle differentiation. Proc Natl Acad Sci U S A. 2002, 99 (17): 11025-11030. 10.1073/pnas.162172899.

    CAS  PubMed  PubMed Central  Google Scholar 

  146. Devarajan E, Sahin AA, Chen JS, Krishnamurthy RR, Aggarwal N, Brun AM, Sapino A, Zhang F, Sharma D, Yang XH, Tora AD, Mehta K: Down-regulation of caspase 3 in breast cancer: a possible mechanism for chemoresistance. Oncogene. 2002, 21 (57): 8843-8851. 10.1038/sj.onc.1206044.

    CAS  PubMed  Google Scholar 

  147. Yoo NJ, Lee JW, Kim YJ, Soung YH, Kim SY, Nam SW, Park WS, Lee JY, Lee SH: Loss of caspase-2, -6 and -7 expression in gastric cancers. APMIS. 2004, 112 (6): 330-335. 10.1111/j.1600-0463.2004.t01-1-apm1120602.x.

    CAS  PubMed  Google Scholar 

  148. Palmerini F, Devilard E, Jarry A, Birg F, Xerri L: Caspase 7 downregulation as an immunohistochemical marker of colonic carcinoma. Hum Pathol. 2001, 32 (5): 461-467. 10.1053/hupa.2001.24328.

    CAS  PubMed  Google Scholar 

  149. Lakhani SA, Masud A, Kuida K, Porter GA, Booth CJ, Mehal WZ, Inayat I, Flavell RA: Caspases 3 and 7: key mediators of mitochondrial events of apoptosis. Science. 2006, 311 (5762): 847-851. 10.1126/science.1115035.

    CAS  PubMed  PubMed Central  Google Scholar 

  150. Gdynia G, Grund K, Eckert A, Bock BC, Funke B, Macher-Goeppinger S, Sieber S, Herold-Mende C, Wiestler B, Wiestler OD, Roth W: Basal caspase activity promotes migration and invasiveness in glioblastoma cells. Mol Cancer Res. 2007, 5 (12): 1232-1240. 10.1158/1541-7786.MCR-07-0343.

    CAS  PubMed  Google Scholar 

  151. Zhou H, Li XM, Meinkoth J, Pittman RN: Akt regulates cell survival and apoptosis at a postmitochondrial level. J Cell Biol. 2000, 151 (3): 483-494. 10.1083/jcb.151.3.483.

    CAS  PubMed  PubMed Central  Google Scholar 

  152. Deshmukh M, Johnson EM: Evidence of a novel event during neuronal death: development of competence-to-die in response to cytoplasmic cytochrome c. Neuron. 1998, 21 (4): 695-705. 10.1016/S0896-6273(00)80587-5.

    CAS  PubMed  Google Scholar 

  153. Martinou I, Desagher S, Eskes R, Antonsson B, Andre E, Fakan S, Martinou JC: The release of cytochrome c from mitochondria during apoptosis of NGF-deprived sympathetic neurons is a reversible event. J Cell Biol. 1999, 144 (5): 883-889. 10.1083/jcb.144.5.883.

    CAS  PubMed  PubMed Central  Google Scholar 

  154. Potts MB, Vaughn AE, McDonough H, Patterson C, Deshmukh M: Reduced Apaf-1 levels in cardiomyocytes engage strict regulation of apoptosis by endogenous XIAP. J Cell Biol. 2005, 171 (6): 925-930. 10.1083/jcb.200504082.

    CAS  PubMed  PubMed Central  Google Scholar 

  155. Potts PR, Singh S, Knezek M, Thompson CB, Deshmukh M: Critical function of endogenous XIAP in regulating caspase activation during sympathetic neuronal apoptosis. J Cell Biol. 2003, 163 (4): 789-799. 10.1083/jcb.200307130.

    CAS  PubMed  PubMed Central  Google Scholar 

  156. Wright KM, Linhoff MW, Potts PR, Deshmukh M: Decreased apoptosome activity with neuronal differentiation sets the threshold for strict IAP regulation of apoptosis. J Cell Biol. 2004, 167 (2): 303-313. 10.1083/jcb.200406073.

    CAS  PubMed  PubMed Central  Google Scholar 

  157. Jia L, Srinivasula SM, Liu FT, Newland AC, Fernandes-Alnemri T, Alnemri ES, Kelsey SM: Apaf-1 protein deficiency confers resistance to cytochrome c-dependent apoptosis in human leukemic cells. Blood. 2001, 98 (2): 414-421. 10.1182/blood.V98.2.414.

    CAS  PubMed  Google Scholar 

  158. Krepela E, Prochazka J, Fiala P, Zatloukal P, Selinger P: Expression of apoptosome pathway-related transcripts in non-small cell lung cancer. J Cancer Res Clin Oncol. 2006, 132 (1): 57-68. 10.1007/s00432-005-0048-6.

    CAS  PubMed  Google Scholar 

  159. Watanabe T, Hirota Y, Arakawa Y, Fujisawa H, Tachibana O, Hasegawa M, Yamashita J, Hayashi Y: Frequent LOH at chromosome 12q22-23 and Apaf-1 inactivation in glioblastoma. Brain Pathol. 2003, 13 (4): 431-439.

    CAS  PubMed  Google Scholar 

  160. Wolf BB, Schuler M, Li W, Eggers-Sedlet B, Lee W, Tailor P, Fitzgerald P, Mills GB, Green DR: Defective cytochrome c-dependent caspase activation in ovarian cancer cell lines due to diminished or absent apoptotic protease activating factor-1 activity. J Biol Chem. 2001, 276 (36): 34244-34251. 10.1074/jbc.M011778200.

    CAS  PubMed  Google Scholar 

  161. Yang L, Mashima T, Sato S, Mochizuki M, Sakamoto H, Yamori T, Oh-Hara T, Tsuruo T: Predominant suppression of apoptosome by inhibitor of apoptosis protein in non-small cell lung cancer H460 cells: therapeutic effect of a novel polyarginine-conjugated Smac peptide. Cancer Res. 2003, 63 (4): 831-837.

    CAS  PubMed  Google Scholar 

  162. Fujita E, Jinbo A, Matuzaki H, Konishi H, Kikkawa U, Momoi T: Akt phosphorylation site found in human caspase-9 is absent in mouse caspase-9. Biochem Biophys Res Commun. 1999, 264 (2): 550-555. 10.1006/bbrc.1999.1387.

    CAS  PubMed  Google Scholar 

  163. Colell A, Ricci JE, Tait S, Milasta S, Maurer U, Bouchier-Hayes L, Fitzgerald P, Guio-Carrion A, Waterhouse NJ, Li CW, Mari B, Barbry P, Newmeyer DD, Beere HM, Green DR: GAPDH and autophagy preserve survival after apoptotic cytochrome c release in the absence of caspase activation. Cell. 2007, 129 (5): 983-997. 10.1016/j.cell.2007.03.045.

    CAS  PubMed  Google Scholar 

  164. Tait SW, Parsons MJ, Llambi F, Bouchier-Hayes L, Connell S, Munoz-Pinedo C, Green DR: Resistance to caspase-independent cell death requires persistence of intact mitochondria. Dev Cell. 2010, 18 (5): 802-813. 10.1016/j.devcel.2010.03.014.

    CAS  PubMed  PubMed Central  Google Scholar 

  165. Chai J, Du C, Wu JW, Kyin S, Wang X, Shi Y: Structural and biochemical basis of apoptotic activation by Smac/DIABLO. Nature. 2000, 406 (6798): 855-862. 10.1038/35022514.

    CAS  PubMed  Google Scholar 

  166. Liu Z, Sun C, Olejniczak ET, Meadows RP, Betz SF, Oost T, Herrmann J, Wu JC, Fesik SW: Structural basis for binding of Smac/DIABLO to the XIAP BIR3 domain. Nature. 2000, 408 (6815): 1004-1008. 10.1038/35050006.

    CAS  PubMed  Google Scholar 

  167. Wu G, Chai J, Suber TL, Wu JW, Du C, Wang X, Shi Y: Structural basis of IAP recognition by Smac/DIABLO. Nature. 2000, 408 (6815): 1008-1012. 10.1038/35050012.

    CAS  PubMed  Google Scholar 

  168. Chen DJ, Huerta S: Smac mimetics as new cancer therapeutics. Anticancer Drugs. 2009, 20 (8): 646-658. 10.1097/CAD.0b013e32832ced78.

    CAS  PubMed  Google Scholar 

  169. Greer RM, Peyton M, Larsen JE, Girard L, Xie Y, Gazdar AF, Harran P, Wang L, Brekken RA, Wang X, Minna JD: SMAC mimetic (JP1201) sensitizes non-small cell lung cancers to multiple chemotherapy agents in an IAP-dependent but TNF-alpha-independent manner. Cancer Res. 2011, 71 (24): 7640-7648. 10.1158/0008-5472.CAN-10-3947.

    CAS  PubMed  PubMed Central  Google Scholar 

  170. Dineen SP, Roland CL, Greer R, Carbon JG, Toombs JE, Gupta P, Bardeesy N, Sun H, Williams N, Minna JD, Brekken RA: Smac mimetic increases chemotherapy response and improves survival in mice with pancreatic cancer. Cancer Res. 2010, 70 (7): 2852-2861. 10.1158/0008-5472.CAN-09-3892.

    CAS  PubMed  PubMed Central  Google Scholar 

  171. Micheau O, Tschopp J: Induction of TNF receptor I-mediated apoptosis via two sequential signaling complexes. Cell. 2003, 114 (2): 181-190. 10.1016/S0092-8674(03)00521-X.

    CAS  PubMed  Google Scholar 

  172. Carter BZ, Mak DH, Morris SJ, Borthakur G, Estey E, Byrd AL, Konopleva M, Kantarjian H, Andreeff M: XIAP antisense oligonucleotide (AEG35156) achieves target knockdown and induces apoptosis preferentially in CD34 + 38- cells in a phase 1/2 study of patients with relapsed/refractory AML. Apoptosis. 2011, 16 (1): 67-74. 10.1007/s10495-010-0545-1.

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We would like to thank everyone in the Chipuk Laboratory for the assistance and support. This work was supported by NIH grant CA157740 (to JEC); a pilot project from NIH P20AA017067 (to JEC), the JJR Foundation (to JEC), the William A. Spivak Fund (to JEC), the Fridolin Charitable Trust (to JEC), and the Developmental Research Pilot Project Program within the Department of Oncological Sciences at Mount Sinai (to JEC). This work was also supported in part by two research grants (5-FY11-74 and 1-FY13-416) from the March of Dimes Foundation (to JEC), and the Developmental Research Pilot Project Program within the Department of Oncological Sciences at Mount Sinai (to JEC).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Jerry E Chipuk.

Additional information

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

RE, TTR, MNS, and JEC discussed and wrote the manuscript. All authors read and approved the final manuscript.

Rana Elkholi, Thibaud T Renault, Madhavika N Serasinghe contributed equally to this work.

Authors’ original submitted files for images

Below are the links to the authors’ original submitted files for images.

Authors’ original file for figure 1

Authors’ original file for figure 2

Authors’ original file for figure 3

Rights and permissions

Open Access  This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made.

The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.

To view a copy of this licence, visit https://creativecommons.org/licenses/by/4.0/.

The Creative Commons Public Domain Dedication waiver (https://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Elkholi, R., Renault, T.T., Serasinghe, M.N. et al. Putting the pieces together: How is the mitochondrial pathway of apoptosis regulated in cancer and chemotherapy?. Cancer Metab 2, 16 (2014). https://doi.org/10.1186/2049-3002-2-16

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/2049-3002-2-16

Keywords