Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Jul 2.
Published in final edited form as: Int Rev Cell Mol Biol. 2020 Jan 27;353:255–284. doi: 10.1016/bs.ircmb.2019.12.005

A connection in life and death: The BCL-2 family coordinates mitochondrial network dynamics and stem cell fate

Megan L Rasmussen a, Vivian Gama a,b,c,d,*
PMCID: PMC7331972  NIHMSID: NIHMS1595427  PMID: 32381177

Abstract

The B cell CLL/lymphoma-2 (BCL-2) family of proteins control the mitochondrial pathway of apoptosis, also known as intrinsic apoptosis. Direct binding between members of the BCL-2 family regulates mitochondrial outer membrane permeabilization (MOMP) after an apoptotic insult. The ability of the cell to sense stress and translate it into a death signal has been a major theme of research for nearly three decades; however, other mechanisms by which the BCL-2 family coordinates cellular homeostasis beyond its role in initiating apoptosis are emerging. One developing area of research is understanding how the BCL-2 family of proteins regulate development using pluripotent stem cells as a model system. Understanding BCL-2 family-mediated regulation of mitochondrial homeostasis in cell death and beyond would uncover new facets of stem cell maintenance and differentiation potential.

Keywords: apoptosis, stem cells, pluripotency, BCL-2 family, mitochondria, mitochondrial dynamics

1. Introduction

Stem cells can simulate the earliest stages of development, since they give rise to the three main tissue lineages (Thomson et al., 1998). This capacity to differentiate into ectoderm, mesoderm, and endoderm lineages is known as pluripotency. Embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs), collectively referred to as PSCs, are the two stem cell types that harbor this ability. Adult stem cells, also known as somatic stem cells, are multipotent and can replenish dying cells in case of tissue damage, and include hematopoietic stem cells, mesenchymal stem cells, and hair follicle stem cells (reviewed in (Goodell et al., 2015)). Stem cells also have the capacity of self-renewal, which is the process by which the stem cell pool is maintained indefinitely. These capabilities to regenerate and to give rise to the three germ layers have propelled an entire field of research dedicated to modeling embryonic development in culture by manipulating key signaling pathways and growth factors. The first human ESC (hESC) line was derived in 1998 from the inner cell mass (ICM) of human blastocysts (Evans and Kaufman, 1981; Martin, 1981; Thomson et al., 1998), while the discoveries of reprogramming mouse and human somatic cells into iPSCs were published in 2006 and 2007, respectively (Takahashi and Yamanaka, 2006; Takahashi et al., 2007). Reprogramming was initially achieved by inducing the expression of master pluripotency transcription factors OCT4 (Octomer-binding transcription factor 4), SOX2 (SRY (sex-determining region Y)-box 2), KLF4 (Kruppel-like factor 4) and c-MYC, collectively known as OSKM), but other methods of attaining iPSCs have been reported (reviewed in (Takahashi and Yamanaka, 2015). The ability of PSCs to self-renew and differentiate has become an efficient tool to study basic processes of human development and various aspects of human diseases, such as diabetes, cardiomyopathy, and cancer (Assady et al., 2001; Hinson et al., 2015; Smith and Tabar, 2019). During embryonic development, genomic instability is especially dangerous for the integrity of rapidly dividing cells of the ICM. Thus, not surprisingly, stem cells are capable of executing intricate programs to quickly respond to apoptotic stress and prevent the propagation of deleterious mutations. Along with the primed cell death program, a growing number of studies on the BCL-2 family have shown changes in mitochondrial dynamics and metabolic function and regulation as stem cells differentiate and as somatic cells reprogram into iPSCs (Rinkenberger et al., 2000; Madden et al., 2011; Prigione et al., 2011; Dumitru et al., 2012; Gama and Deshmukh, 2012; Rasmussen et al., 2018). In the following chapter, we will discuss the known fundamental mechanisms involved in these changes, centering on the BCL-2 family, as well as describe areas that are open to more detailed exploration (Figure 1). In addition, many aspects of mitochondrial biology are beginning to emerge as hallmarks of pluripotency and self-renewal (Wanet et al., 2015; Rastogi et al., 2019). The increased sensitivity to apoptosis, the changes in mitochondrial morphology and localization, and the shifting of the metabolic program all accompany reprogramming. Furthermore, cellular events such as mitochondrial biogenesis, mitochondrial trafficking and motility, and mitochondrial DNA (mtDNA) transcription could also be important for reprogramming and generation of specialized tissues. Thus, the unique properties of ESCs and iPSCs make them a valuable model system to illuminate the effects of these processes on self-renewal and differentiation.

Figure 1: The BCL-2 family regulates mitochondrial cell death and homeostasis in stem cells.

Figure 1:

This schematic depicts the canonical pathways of mitochondrial apoptosis and priming. Highlighted are the reported changes in PSC regulation of these pathways: 1) High levels of pro-apoptotic proteins. 2) BAX is maintained in an active state at the Golgi. 3) High levels of MCL-1, which is important for pluripotent maintenance and mitochondrial fission. 4) Increased fragmentation of the mitochondrial network and higher dependence on glycolytic metabolism.

2. The BCL-2 family in stem cell death

2.1. Mitochondrial pathway of apoptosis

Caspase-dependent apoptosis occurs through both extrinsic and intrinsic pathways, which are mediated by external death ligands and mitochondrial-localized proteins, respectively (Elmore, 2007). The focus of this chapter will be on the intrinsic or mitochondrial pathway of apoptosis; the extrinsic apoptotic pathway is another form of regulated cell death that depends on detection and propagation of extracellular signals, which has been comprehensively reviewed here (Ashkenazi and Dixit, 1998; Mehlen and Bredesen, 2011; Galluzzi et al., 2018). The intrinsic pathway of apoptosis is crucial for embryonic development, tissue homeostasis, and in cellular response to irreversible perturbations. When the proteins that regulate apoptosis are dysregulated, cells can become cancerous and contribute to tumor formation, or die uncontrollably resulting in neurodegeneration. The BCL-2 family are the main regulatory proteins that control the intrinsic pathway of cell death (Figure 2). The direct binding interactions that occur between pro-apoptotic and anti-apoptotic family members govern their activities, ultimately resulting in MOMP after receiving a death signal (Oltval et al., 1993; Kale et al., 2018). Roughly 30 family members have been identified to belong to the BCL-2 family, which is made up of structurally similar globular proteins that are defined by multiple domains, known as BCL-2 homology (BH) domains (BH1, BH2, BH3, and BH4) (Tsujimoto et al., 1984; Wang et al., 1996; Yang et al., 1996; Lindsten et al., 2000; Chipuk et al., 2010). Family members with multiple BH domains are either pro-apoptotic “effectors” (BCL-2 associated X (BAX), BCL-2 antagonist killer 1 (BAK1; known as BAK), BCL-2 ovarian killer (BOK)) or anti-apoptotic/pro-survival (BCL-2 apoptosis regulator (BCL-2), BCL-2-like 1 (known as BCL-xL), BCL-2-like 2 (BCL-w), Myeloid cell leukemia (MCL-1), and BCL-2related protein A1 (BFL-1/A1)). Another group of family members have only one BH domain, known as the BH3-only proteins. The “activator” pro-apoptotic proteins (BCL-2 interacting mediator of cell death (BIM), BCL-2 interacting domain death agonist (BID), P53-upregulated modulator of apoptosis (PUMA)) and “sensitizer” pro-apoptotic proteins (phorbol-12-myristate-13-acetate-induced protein 1 (PMAIP1, best known as NOXA), BCL-2-associated agonist of cell death (BAD), BCL-2 interacting killer (BIK), BCL-2 modifying factor (BMF), and Harakiri (HRK)) work to either activate the effector proteins directly or sequester the anti-apoptotic family members from binding the effectors. After activation of BAK, BAX and/or BOK by the BH3-only proteins, they oligomerize to form pores in the outer mitochondrial membrane (OMM), initiating MOMP, reducing mitochondrial membrane potential and releasing cytochrome c from the inner membrane space (IMS) (Liu et al., 1996; Lindsten et al., 2000; Goldstein et al., 2000; Eskes et al., 2000; Kalkavan and Green, 2018; Moldoveanu and Czabotar, 2019). Mechanistically, each of the effector proteins are regulated through slightly different means. While BAK is usually found anchored to the OMM, BAX resides in the cytosol in an inactive state and must undergo a conformational change to insert into the OMM through its C-terminal domain (Hsu and Youle, 1997; Wolter et al., 1997; Goping et al., 1998; Gavathiotis et al., 2008; Green and Chipuk, 2008; Todt et al., 2015). Less is known about BOK activity and regulation, but it is proposed to have the ability to initiate MOMP independent of BAK or BAX, and it is modulated by the endoplasmic-reticulum-associated protein degradation (ERAD) pathway (Llambi et al., 2016; Schulman et al., 2016; Zheng et al., 2018; Schulman et al., 2019). Cytochrome c release results in Apoptotic protease activating factor-1 (APAF-1)/Caspase-9 assembly, known as the apoptosome (Zou et al., 1997; Kim et al., 2005; Taylor et al., 2008; Inoue et al., 2009), which activates the executioner caspases, leading to the extensive cleavage and destruction of critical cellular components (Li et al., 1997; Dix et al., 2008; Mahrus et al., 2008; Fuchs and Steller, 2011; Ramirez and Salvesen, 2018). Morphological changes occur, including cell constriction, organelle fragmentation, chromatin condensation, plasma membrane “blebbing” as the cortex ruptures, and finally the break-up of the cell into apoptotic bodies, which are the defining features of apoptosis (Kerr et al., 1972).

Figure 2: The BCL-2 family regulates the mitochondrial pathway of apoptosis.

Figure 2:

When cells receive an intrinsic death signal, BAX becomes activated by the BH3-only activators, undergoes a conformational change, and translocates to the outer mitochondrial membrane (OMM). BAX (in addition to BAK and BOK) then oligomerize and form pores in the OMM, causing MOMP and the release of cytochrome c into the cytosol. BAX activation can be prevented by sequestration of the activator BH3-only proteins through the pro-survival proteins BCL-2, MCL-1, BCL-xL, etc, which are inhibited by the sensitizer BH3-only family members. In human embryonic stem cells, BAX can be maintained in an active state at the Golgi, which confers quick translocation to the OMM in case of a death signal. Additionally, MCL-1 has been found to reside at the matrix in stem cells, where it is proposed to interact with OPA1.

There are several proposed models of how the BCL-2 family execute cell death upstream of MOMP (reviewed in (Shamas-Din et al., 2013)): the direct activation model, the displacement model, the embedded together model, and the unified model. The latter two models, which built upon the direction activation and displacement models, highlight the complexity of the mitochondrial cell death pathway. The embedded together model proposes that the specific interactions of the BCL-2 family members are dependent on cellular equilibria, with binding events occurring based on concentration and binding affinities of each protein (Leber et al., 2007). These affinities are also dependent on proximity to the mitochondrial membrane as well as post-translational modifications. The unified model of apoptosis differs in that it proposes that the anti-apoptotic proteins have two “modes”: one in binding and sequestering the activator BH3-only proteins, which can be overcome by the sensitizer BH3-only proteins, and the other binding and sequestering BAK/BAX, which results in a more robust inhibition of MOMP (Llambi et al., 2011; Kalkavan and Green, 2018). The unified model also includes the effects that the BCL-2 family proteins have on mitochondrial dynamics, thus expanding on the embedded together model.

2.2. The BCL-2 family in stem cells

The intrinsic apoptosis has been shown to be uniquely regulated in stem cells (Madden et al., 2011; Dumitru et al., 2012; Gama and Deshmukh, 2012; Liu et al., 2013; Rasmussen et al., 2018). Human PSCs (hPSCs) are extremely sensitive to DNA perturbations in a P53-dependent manner. P53 protein expression, however, was not significantly different between hPSCs and differentiated cells, pointing to another mode of action for this rapid apoptotic response. It was found that expression of the BCL-2 family in stem cells is shifted so that pro-apoptotic members are upregulated, while pro-survival members are downregulated, which serves to position them closer to the threshold of apoptosis, a property known as mitochondrial priming (Certo et al., 2006; Liu et al., 2013). BAX aides in this high mitochondrial priming state of hPSCs (Dumitru et al., 2012). BAX was found in an activated state in homeostatic conditions but localized at the Golgi until an apoptosis-initiating event occurred (e.g. etoposide-mediated DNA damage), when it would translocate to the mitochondria (Dumitru et al., 2012). How BAX is maintained at the Golgi in an active state remains elusive, as up to this point active BAX, which is identified by its N-terminal domain (Upton et al., 2007), had only been known to localize at the OMM, and furthermore only in dying cells. When hPSCs were differentiated into stem cell aggregates known as embryoid bodies, BAX was no longer active during homeostasis and retained its mitochondrial localization during apoptotic stress, indicating a stem cell-specific mechanism of BCL-2 family regulation (Dumitru et al., 2012). Maintaining BAX in an active conformation and in a high mitochondrial priming state likely allows for hPSCs to respond rapidly to cell death, thereby preventing potential mutations induced by DNA damage from persisting in the stem cell pool or in the differentiating cell lineage. Conversely, sustaining BAX in an active state would also pose significant risk for spontaneous apoptosis. Thus, isolating BAX at the Golgi could serve to safeguard hPSCs from unnecessary cell death while still allowing for its rapid induction if the cell is antagonized. At the organismal level, this sensitive response may stop the development of abnormal cells in the growing embryo. If constitutive BAX activation is also maintained by other highly proliferative cells in adult tissue, then perhaps it prevents aberrant growth of cancer cells and tumors. Dissection of the mechanisms underpinning BAX activation and localization, both at the Golgi and at the mitochondria, will be necessary to fully elucidate this apoptotic sensitivity as a defining component of stem cell homeostasis. As PSCs begin to differentiate, they quickly become resistant to DNA damage-mediated apoptosis; this was shown to be facilitated in part by a decrease in mitochondrial priming (Madden et al., 2011; Liu et al., 2013). Mitochondrial priming and apoptotic sensitivity have been exploited in recent years to identify dependencies of cancer cells on specific BCL-2 family proteins. This technique is known as BH3 profiling (Certo et al., 2006; Montero et al., 2015; Montero and Letai, 2016), and it is useful to measure the different binding affinities between the BCL-2 family members across cell types and patient-derived tumors.

The pro-apoptotic members of the BCL-2 family become activated through several upstream sensing mechanisms. As previously mentioned, PSCs are highly sensitive to DNA damage, which in turn initiates the transcription factor P53 and triggers a repair response and cell cycle arrest (Kruse and Gu, 2009). If the cell cannot resolve the damage, the BCL-2 family facilitates MOMP and the cell undergoes apoptosis in a caspase-dependent manner. Fluctuations in P53 activation have been shown to determine cell fate (Purvis et al., 2012). During reprogramming of somatic cells back to a pluripotent state, inhibition of P53 greatly improves cell survival due to the bypass of BCL-2 family-mediated cell death (Kawamura et al., 2009; Li et al., 2013). Excellent reviews covering the details of P53 function in stem cells can be found here (Bonizzi et al., 2012; Levine et al., 2016; Jain and Barton, 2018). Downstream of BCL-2 family facilitation of MOMP, activity of caspases has been shown to play dual roles in both promoting differentiation and maintaining proper distribution of differentiated cell types, as well as driving reprogramming of somatic cells (Fujita et al., 2008; Li et al., 2010; Fu et al., 2019).

3. The BCL-2 family in mitochondrial dynamics

The main players of intrinsic apoptosis, the BCL-2 family of proteins, execute their functions at the mitochondria. Beyond being the energy-producing powerhouses of the cell, mitochondria serve as major signaling organelles and constantly fuse and divide in a process known as mitochondrial dynamics (Chan, 2007; Berman et al., 2008; Friedman and Nunnari, 2014) (Figure 3). These continuous, energy-consuming events are proposed to occur both in response to various stimuli and to relay signals throughout the cell to other organelles. Dynamic movement and fragmentation of the mitochondrial network was first documented in 1915, with various morphological changes described in detailed, hand-drawn illustrations (Lewis and Lewis, 1914). Yet, the notion that mitochondria are static, bean-like organelles persevered over the decades. The development of more advanced imaging technologies has allowed for improved observation and tracking of mitochondrial dynamics and motility, which has propelled the field and revealed the remarkable changes that mitochondria undergo within cells (Johnson et al., 1981; Bereiter-Hahn and Vöth, 1994; Rizzuto et al., 1996). Emerging studies have also highlighted new links between mitochondrial dynamics machinery, apoptosis and mitochondrial autophagy (mitophagy) (Kageyama et al., 2014; Morciano et al., 2016b; Rasmussen et al., 2018; Fu et al., 2019). Proper equilibrium of mitochondrial dynamics is essential for cellular and organismal homeostasis, and diseases often manifest when the proteins that regulate fission and fusion are mutated (Chan, 2007; Westermann, 2010; Dorn, 2013; Chan, 2019).

Figure 3: The dynamic cycle of mitochondrial morphology and quality control.

Figure 3:

In homeostasis, differentiation, reprogramming, and during stress, the mitochondrial network undergoes rounds of fusion and fission. During fission, DRP-1 is activated and recruited to pre-constricted regions of mitochondria (facilitated by actin and ER tubules) by its receptors (MFF, MID49/51, FIS1). Mitochondrial fusion is achieved by the activity of MFNs, which dimerize with MFNs on adjacent mitochondria to fuse the OMM. This is followed by OPA1-mediated fusion of the inner mitochondria membrane (IMM). If mitochondria are damaged (i.e. low membrane potential), PINK1 is stabilized at the mitochondria and phosphorylates proteins on the OMM. This phosphorylation triggers Parkin-mediated ubiquitination of OMM proteins, causing recruitment of various adapter proteins and receptors associated with the phagophore. The damaged mitochondrion is then engulfed by the mature mitophagophore, which fuses with the lysosome for degradation.

3.1. Mitochondrial fission

Mitochondrial shape changes drastically depending on the cell type, cell cycle stage, and metabolic state (Chen and Chan, 2017). Differentiated cell types, such as fibroblasts, myocytes, and endothelial cells, generally have elongated, cristae-rich mitochondria that make up complex networks, which promote mtDNA homogenization and energy generation (Kuznetsov et al., 2009). Mitochondrial fission is necessary for distribution of mitochondria to daughter cells during cell division, mitochondrial transport within cells, and isolation of damaged mitochondria for degradation by mitophagy (Nunnari et al., 1997; Pon, 2013; Ganesan et al., 2019). As differentiated cells reprogram and transition into a stem-like state, mitochondria undergo higher levels of fission and arrange to be more perinuclear (Prieto et al., 2016; Chen and Chan, 2017). The mitochondria of hESCs and iPSCs are considered more functionally immature, with globular shapes and fewer cristae, contributing to a metabolic profile that is traditionally thought to be more dependent on glycolysis for energy requirements (Facucho-Oliveira and St. John, 2009; Chung et al., 2010). Constitutive activation of the dynamin-related guanosine triphosphates (GTPases) that control fission is likely a major participating factor in maintaining high levels of fragmentation in stem cells (Chung et al., 2010; Prieto et al., 2016; Rasmussen et al., 2018).

The large dynamin-related GTPases regulate mitochondrial dynamics by inducing mitochondrial fission (division) and fusion (reviewed in section 3.2) in a highly conserved manner (Praefcke and McMahon, 2004; Hoppins et al., 2007; Chan, 2012). Dynamin-related protein 1 (DRP-1) executes its function upon activation by several post-translational modifications, including phosphorylation, ubiquitination, sumoylation, and O-GlcNAcylation (Westermann, 2010). The enzymes that perform this activation of Dynamin-related proteins have not been completely identified, but ERK1/2 and CDK1 have been shown phosphorylate DRP-1 at Serine 616, providing insight into specific pathways that promote increased mitochondrial fragmentation (Taguchi et al., 2007; Prieto et al., 2016). Once DRP-1 is activated, it translocates from the cytosol to the OMM, where it hydrolyzes GTP and self-assembles around the mitochondria, constricting both membranes until the organelle is divided in two (Hoppins et al., 2007; Pon, 2013; Antonny et al., 2016; Francy et al., 2017). The structural domains and mechanistic details of action for DRP-1 have been described in several studies and reviews (Hoppins et al., 2007; Meglei and McQuibban, 2009; Chappie et al., 2010; Gao et al., 2010; van der Bliek and Payne, 2010; Mears et al., 2011; Francy et al., 2017); however, whether or not DRP-1 activation is differentially regulated in PSCs, which have a more fragmented mitochondrial network, has not been elucidated. DRP-1 function in mitochondrial fission is required for proper achievement of pluripotency early on in the reprogramming process (Prieto et al., 2016). As DRP-1 is also involved in the mitochondrial fragmentation observed during apoptosis (Frank et al., 2001), and iPSCs are highly sensitive to apoptotic stimuli, these cells are likely highly dependent on DRP-1 beyond its role in homeostatic conditions. It is important to note that some studies suggest the possibility that mitochondrial fission can occur in the absence of DRP-1 (Onoue et al., 2013; Stavru et al., 2013), and/or that DRP-1 requires additional proteins for severance of the mitochondria (i.e. dynamin-2) (Lee et al., 2016), a phenomenon that remains controversial (Kamerkar et al., 2018; Fonseca et al., 2019). Moving forward, additional studies into the mechanisms by which mitochondrial fission is regulated across different cell types, cell states and model systems are needed for appropriately interpreting conflicting results, and to better elucidate the intricate events coordinating mitochondrial fragmentation.

3.2. Mitochondrial fusion

As stem cells differentiate and become more mature, the mitochondrial network transforms to support the changing needs of the differentiating cell. Mitochondrial fusion is required for the proper maturation of several tissue types, and it is necessary for mtDNA homogenization and assembly of the electron transport chain (ETC) (Chan, 2012). The specific mechanisms by which proteins control mitochondrial fusion are not well understood. In vitro studies have shown that OMM fusion is a mechanistically distinct process from fusion of the inner mitochondrial membrane (IMM) (Legros et al., 2002; Meeusen et al., 2004). The large GTPases that govern the process of mitochondrial fusion are Mitofusin 1 (MFN1), Mitofusin 2 (MFN2), and Optic Atrophy 1 (OPA1) (Alexander et al., 2000; Delettre et al., 2000; Chen et al., 2003). MFN1 and MFN2 are localized to the OMM, where they facilitate outer membrane fusion by homo- or hetero-dimerizing with MFNs on adjacent mitochondria and actively fusing the outer membranes. While the MFN paralogs are highly similar, they are not completely redundant and are both required for mitochondrial fusion (Escobar-Henriques and Joaquim, 2019). Depletion of MFN1 in mouse embryonic fibroblasts (MEFs) leads to increased fragmentation of individual mitochondria, resulting in small fragments, while MFN2 depletion results in larger fragments of aggregated mitochondria (Chen et al., 2003). Interestingly, double knockout embryos die earlier than single mutants (Chen et al., 2003) and both MFN1 and 2 levels vary in expression among different tissues (Santel et al., 2003), pointing to a non-redundant requirement for the Mitofusins during early development (Schrepfer and Scorrano, 2016).

Once OMM fusion occurs, the IMM is fused through the activity of OPA1. OPA1 regulation is complex, as it has eight different splice isoforms that lead to the formation of two proteolytically cleaved proteins, designated as long OPA1 (OPA1-L) and short OPA1 (OPA1-S), which serve several distinct and overlapping purposes (Mishra et al., 2014; Del Dotto et al., 2017). OPA1-L is anchored in the IMM with its GTPase domain exposed to IMS. It coordinates the active process of IMM fusion by forming homo-dimers with OPA1-L proteins on the opposite target membrane. OPA-1S is proposed to provide a more passive, structural role at the mitochondrial matrix in cristae formation and maintenance, while combinations of both long and short forms work together to fine-tune mitochondrial morphology and function (Del Dotto et al., 2018). The OPA1-S form has also been implicated in fission of the IMM (Anand et al., 2014). These studies were performed in MEFs, but it would be interesting to probe if the homeostatic balance of OPA1 isoforms is differentially regulated in stem cells and over the course of differentiation. Disproportionate levels of either mitochondrial fusion or fission results in profound pathological abnormalities. These include embryonic lethality at mid-gestation in mice deficient in any of the dynamin-related proteins (i.e. MFN1, MFN2, OPA1, and DRP1) (Chen et al., 2003; Davies et al., 2007; Alavi et al., 2007; Ishihara et al., 2009; MacVicar and Langer, 2016), as well as neurodegenerative diseases such as Charcot-Marie-Tooth syndrome and dominant optic atrophy (Züchner et al., 2004; Alavi et al., 2007; Davies et al., 2007; Waterham et al., 2007) caused by mutations in MFN2 and OPA1, respectively.

3.3. Mitochondrial dynamics and apoptosis

Mitochondrial dynamics also change during apoptosis; mitochondrial fusion is thought to protect against mitochondrial pore formation (Estaquier and Arnoult, 2007; Jahani-Asl et al., 2010), while mitochondrial fission has been shown occur concurrently with BAK/BAX-mediated cell death (Frank et al., 2001; Youle and van der Bliek, 2012). Parallel to this, studies have shown that pro-apoptotic BAK and BAX must be inactivated for mitochondrial fusion to occur, and BAX has been shown to modulate fusion through interactions with the Mitofusins (Karbowski et al., 2006; Brooks et al., 2007; Hoppins et al., 2007, 2011). Additionally, other BCL-2 family members have recently been associated with the maintenance of mitochondrial dynamics in various cell types (Chen et al., 2011; Hardwick and Soane, 2013). In adult neurons, the anti-apoptotic protein BCL-xL is enriched in the mitochondria, where it promotes proper mitochondrial length and size, as well as localization of the mitochondria to the highly energetic synapses (Berman et al., 2008; Li et al., 2008). BCL-xL has been reported to be involved in neuronal connectivity and communication, which are required for normal brain development (Nakamura et al., 2016). Another anti-apoptotic family member, MCL-1, has been shown to also function in the regulation of mitochondrial dynamics in cancer cells and hPSCs (Morciano et al., 2016a; Rasmussen et al., 2018). MCL-1 is expressed at high levels in hPSCs, and its depletion causes mitochondrial elongation in stem cells that coincides with the loss of expression of pluripotency markers OCT4 and NANOG (Rasmussen et al., 2018). The changes in mitochondrial morphology are likely due to MCL-1’s interaction with DRP-1, but the details of the state of DRP-1 activation are still unknown. Interestingly, MCL-1 was also shown to interact with OPA1, an association that likely occurs through MCL-1’s matrix-localized form (Rasmussen et al., 2018). These non-apoptotic interactions further implicate MCL-1, and perhaps other BH domain proteins, in the modulation of mitochondrial dynamics and homeostasis beyond their roles in cell death. A recent study showed that BID, a pro-apoptotic BH3-only protein, is also imported into the mitochondrial matrix where it regulates cristae morphology and organization (Salisbury-Ruf et al., 2018). As BID is a known binding protein of MCL-1, this study showed these two family members interact at the matrix in cancer cells. It would be interesting to elucidate the dynamics of this interaction in stem cells. For instance, MCL-1 deficiency causes significant changes in the mitochondrial network of iPSCs where BID is highly expressed (Rasmussen et al., 2018). Whether MCL-1’s remodeling of the mitochondria is dependent on BID or whether MCL-1 and BID work in concert at the matrix to maintain pluripotency/self-renewal properties in stem cells is not known. Understanding the regulation of cell fate from the perspective of mitochondrial dynamics and the BCL-2 family of proteins could shed light on the intricacies of cell signaling and organelle biology.

As previously mentioned, extensive remodeling of the mitochondrial network occurs during apoptosis. These changes result in the release of cytochrome c and other caspase-inducing proteins from the IMS (Goldstein et al., 2000; Garrido et al., 2006; Galluzzi et al., 2018). Mitochondrial fragmentation during apoptosis takes place through two synchronized, but independent, events: dissociation of cristae junctions, where pools of cytochrome c are located, and BAK/BAX oligomerization and pore formation at the outer membrane (Gao et al., 2001; Lee et al., 2004; Ow et al., 2008; Sheridan et al., 2008; Montessuit et al., 2010; Sinibaldi et al., 2013). A growing number of studies show that DRP-1 acts in response to apoptotic signals, consistent with the idea that mitochondrial fission occurs along with MOMP. DRP-1 has been shown promote BAX translocation to the mitochondria (Wang et al., 2015), and it also localizes to BAX/BAK pores (Frank et al., 2001; Tanaka and Youle, 2008) where it promotes extensive mitochondrial network fission. DRP-1 depletion prevents mitochondrial fission during apoptosis, and overexpression of the DRP-1 mutant K38A, which inhibits GTP binding by DRP-1, prevents apoptosis-induced mitochondrial fission (Smirnova et al., 2001; Sugioka et al., 2004; Estaquier and Arnoult, 2007). BAK and BAX have also been reported to localize at fission sites along with DRP-1 and MFN2, providing further evidence that MOMP is associated with the fission regulators (Karbowski et al., 2002; Hoppins et al., 2011).

OPA1 has been shown to be involved during MOMP where its disassembly is needed to facilitate cytochrome c release (Frezza et al., 2006; Yamaguchi et al., 2008; Jiang et al., 2014). A number of studies demonstrated that apoptosis requires OPA1-dependent cristae remodeling initiated by metalloprotease OMA1-mediated cleavage of OPA1. Furthermore, activation of OMA1 is mediated by pro-apoptotic members of the BCL-2 family. How these functions of OPA1 are regulated during apoptosis in stem cells is not clear, however, these studies point to distinct functions of OPA1 in mitochondrial fusion and in cristae remodeling during apoptosis (Arnoult et al., 2005; Frezza et al., 2006; Yamaguchi et al., 2008; Jiang et al., 2014).

3.4. Mitophagy

Concomitant with increased mitochondrial fission is the clearance of damaged mitochondria by mitophagy (Zhang, 2013; Burman et al., 2017) (Figure 3). Mitophagy is proposed to be an opposing process to apoptosis, and initiation of either pathway is dependent on signaling crosstalk and cell state (Pickles et al., 2018). Activation of PTEN-induced putative kinase 1 (PINK1) aids in the selective clearance of mitochondria through recruitment of the E3 ubiquitin ligase Parkin, which translocates to the mitochondria from the cytosol and targets mitochondrial membrane proteins for degradation by the autophagosome (Narendra et al., 2008). MFN1 and MFN2 are substrates for Parkin-mediated ubiquitination (Gegg et al., 2010). However, this modification targets them for proteasomal degradation rather than acting as a mitophagy signal, suggesting that elimination of mitochondrial fusion and a shift to fission occurs prior to mitophagy. BCL-2 and BCL-xL have been shown to negatively regulate mitophagy through inhibition of Beclin-1 (also known as ATG6), a BH3 domain-containing protein that regulates formation of the phagophore along with adaptor proteins such as BNIP3 (BCL2 and adenovirus E1B 19 kDa-interacting protein 3) and NIX (BNIP3-like) (Pattingre et al., 2005; Maiuri et al., 2007; Zhang and Ney, 2009; Dhingra et al., 2014; Vásquez-Trincado et al., 2016; Fernández et al., 2018). Another study in HeLa cells showed that overexpression of any of the anti-apoptotic family members (BCL-2, BCL-xL, BCL-w, MCL-1, A1) prevented PINK1/Parkin-mediated mitophagy, but this block was independent of Beclin-1 (Hollville et al., 2014).

Mitophagy can maintain cellular homeostasis and is also triggered by nutrient deprivation and other stressors; however, mitophagy can also be developmentally initiated (Pickles et al., 2018). In fact, decreased levels of mitophagy accompany increased mitochondrial fusion that occurs during stem cell differentiation, and increased mitophagy during somatic cell reprogramming promotes the switch in metabolism back to glycolytic dependence, as it reduces mitochondrial mass, mtDNA content, and ETC components that facilitate oxidative phosphorylation (Vazquez-Martin et al., 2012; Fu et al., 2019). Mitophagy is essential in post-mitotic cells, since they have lost their proliferative capacity and rely on maintaining a healthy pool of mitochondria (Zhang, 2013; Evans and Holzbaur, 2019). However, high levels of mitochondrial fission in stem cells would perhaps require high activation of mitophagy for quality control purposes (Naik et al., 2019). Supporting this, late-passage PINK1-deficient mouse iPSCs, while they expressed the pluripotency factors OCT4 and SOX2, were more prone to differentiation and showed accumulation of damaged mitochondria (Vazquez-Martin et al., 2016).

3.5. Inter-organelle contacts

Although it is known that mitochondrial fragmentation and the protein machinery that coordinate this process are essential, details of how mitochondrial membrane integrity is disrupted, yet in a controlled manner, are not well understood. Structural analysis of DRP-1 discovered that the rings formed by DRP-1 oligomers are smaller in diameter (30–50 nm) than the average diameter of mitochondria (Ingerman et al., 2005; Mears et al., 2011), suggesting there are likely other constriction mechanisms involved. Recent work has identified the endoplasmic reticulum (ER) as well as the actin cytoskeleton as key players in mitochondrial dynamics (Lewis et al., 2016; Rehklau et al., 2017; Steffen and Koehler, 2018). ER tubules can be found at sites of fission, where they form contacts with the mitochondria through the ER-mitochondria encounter structure (ERMES) complex and initiated fission even before DRP-1 constriction (Elgass et al., 2015; Lewis et al., 2016; Yang and Svitkina, 2019). Tethering of the ER to mitochondria might serve not only to facilitate mitochondrial fission, but also to regulate mitochondrial membrane biosynthesis, calcium signaling, and protein import (reviewed in (Kornmann and Walter, 2010)). Following ER tubule encirclement of the mitochondria, it is proposed that INF2-induced actin polymerization occurs, triggering an initial constriction mechanism to facilitate DRP-1 recruitment, assembly, and final scission of the membrane (Korobova et al., 2013; Pon, 2013). It will be interesting to determine if members of the BCL-2 family such as MCL-1 and BAK/BAX are involved in this mitochondrial fission “complex” in cells subjected to sublethal doses of apoptotic stimuli. It could also be informative to examine potential adaptations to these processes in hPSCs and cancer stem cells where the balance of mitochondrial dynamics is shifted to favor high fission activity.

Increased mitochondrial priming and mitochondrial fission are two vital components that allow for pluripotent maintenance, and these properties change upon cellular differentiation; however, the precise signaling events and protein interactions that govern these processes are yet to be elucidated. The BCL-2 family affects mitochondrial priming, but we also propose that BCL-2 proteins regulate fragmentation; reprogramming of differentiated cells into iPSCs will be a crucial tool to dissect this intriguing possibility in a temporal and cell-specific manner. Further investigation into the relationship between mitochondrial priming, fragmentation and the acquisition of pluripotency or a particular stem cell fate opens an exciting opportunity for future mechanistic studies.

4. Concluding remarks and future perspectives

As discussed throughout this chapter, the BCL-2 family of proteins regulate the mitochondrial pathway of apoptosis and are thereby at the center of several essential signaling pathways. The mitochondrial network is responsible for the regulation of apoptosis, oxidative phosphorylation, coordination of neighboring organelles, and they provide the machinery needed to generate key metabolites that serve the bioenergetic and biosynthetic needs of the cell (Fu et al., 2019). The crosstalk between the pathways of BCL-2 family-mediated cell death, mitochondrial dynamics, and mitophagy must be explored further in PSCs, as it will likely provide important details into how cells navigate the differentiation and reprogramming processes. The role of reactive oxygen species on stem cell maintenance is also an intriguing area of study, since multiple redox-sensitive pathways also control apoptosis and mitochondrial dynamics (e.g. MAPK, ERK1/2, JNK) (Bigarella et al., 2014).

As discussed in this chapter, many questions remain; for example: 1) How do the signaling pathways that converge at the mitochondria intersect to regulate cell fate? 2) How are the changes in mitochondrial network, mitochondrial turnover, and mitochondrial connectivity to other organelles regulated as cells undergo differentiation and reprogramming? 3) How do changes in mitochondrial morphology contribute to adaptations in metabolic state? 4) How do metabolite levels modulate the activity of epigenetic enzymes ultimately affecting gene expression? Answering these questions would open the opportunity to advance the fields of stem cell biology and apoptosis as well as to rationalize new therapies, as shown by the emergence of pharmacological inhibitors of the BCL-2 family.

Box 1: Abbreviations used in this chapter.

graphic file with name nihms-1595427-f0004.jpg

Acknowledgments

We apologize to colleagues whose work has not been included in this review due to space limitations. We would like to thank the Gama laboratory for their experimental and conceptual contributions, which led to some of the ideas presented in this review.

This work was supported by NIH Grants: 1R35GM128915-01 (to VG), 1R21CA227483-01A1 (to VG), and AHA Grant: 19PRE34380515 (to MR).

References

  1. Alavi MV, Bette S, Schimpf S, Schuettauf F, Schraermeyer U, Wehrl HF, Ruttiger L, Beck SC, Tonagel F, Pichler BJ, Knipper M, Peters T, Laufs J, Wissinger B, 2007. A splice site mutation in the murine Opa1 gene features pathology of autosomal dominant optic atrophy. Brain 130, 1029–1042. 10.1093/brain/awm005 [DOI] [PubMed] [Google Scholar]
  2. Alexander C, Votruba M, Pesch UEA, Thiselton DL, Mayer S, Moore A, Rodriguez M, Kellner U, Leo-Kottler B, Auburger G, Bhattacharya SS, Wissinger B, 2000. OPA1, encoding a dynamin-related GTPase, is mutated in autosomal dominant optic atrophy linked to chromosome 3q28. Nat. Genet 26, 211–215. 10.1038/79944 [DOI] [PubMed] [Google Scholar]
  3. Anand R, Wai T, Baker MJ, Kladt N, Schauss AC, Rugarli E, Langer T, 2014. The i-AAA protease YME1L and OMA1 cleave OPA1 to balance mitochondrial fusion and fission. J. Cell Biol 204, 919–929. 10.1083/jcb.201308006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Antonny B, Burd C, De Camilli P, Chen E, Daumke O, Faelber K, Ford M, Frolov VA, Frost A, Hinshaw JE, Kirchhausen T, Kozlov MM, Lenz M, Low HH, McMahon H, Merrifield C, Pollard TD, Robinson PJ, Roux A, Schmid S, 2016. Membrane fission by dynamin: what we know and what we need to know. EMBO J. 35, 2270–2284. 10.15252/embj.201694613 [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Arnoult D, Grodet A, Lee Y-J, Estaquier J, Blackstone C, 2005. Release of OPA1 during Apoptosis Participates in the Rapid and Complete Release of Cytochrome c and Subsequent Mitochondrial Fragmentation. J. Biol. Chem 280, 35742–35750. 10.1074/jbc.M505970200 [DOI] [PubMed] [Google Scholar]
  6. Ashkenazi A, Dixit VM, 1998. Death Receptors: Signaling and Modulation. Science 281, 1305–1308. 10.1126/science.281.5381.1305 [DOI] [PubMed] [Google Scholar]
  7. Assady S, Maor G, Amit M, Itskovitz-Eldor J, Skorecki KL, Tzukerman M, 2001. Insulin Production by Human Embryonic Stem Cells. Diabetes 50, 1691–1697. 10.2337/diabetes.50.8.1691 [DOI] [PubMed] [Google Scholar]
  8. Bereiter-Hahn J, Vöth M, 1994. Dynamics of mitochondria in living cells: Shape changes, dislocations, fusion, and fission of mitochondria. Microsc. Res. Tech 27, 198–219. 10.1002/jemt.1070270303 [DOI] [PubMed] [Google Scholar]
  9. Berman SB, Pineda FJ, Hardwick JM, 2008. Mitochondrial fission and fusion dynamics: the long and short of it. Cell Death Differ. 15, 1147–1152. 10.1038/cdd.2008.57 [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Bigarella CL, Liang R, Ghaffari S, 2014. Stem cells and the impact of ROS signaling. Development 141, 4206–4218. 10.1242/dev.107086 [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Bonizzi G, Cicalese A, Insinga A, Pelicci PG, 2012. The emerging role of p53 in stem cells. Trends Mol. Med 18, 6–12. 10.1016/j.molmed.2011.08.002 [DOI] [PubMed] [Google Scholar]
  12. Brooks C, Wei Q, Feng L, Dong G, Tao Y, Mei L, Xie Z-J, Dong Z, 2007. Bak regulates mitochondrial morphology and pathology during apoptosis by interacting with mitofusins. Proc. Natl. Acad. Sci 104, 11649–11654. 10.1073/pnas.0703976104 [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Burman JL, Pickles S, Wang C, Sekine S, Vargas JNS, Zhang Z, Youle AM, Nezich CL, Wu X, Hammer JA, Youle RJ, 2017. Mitochondrial fission facilitates the selective mitophagy of protein aggregates. J. Cell Biol 216, 3231–3247. 10.1083/jcb.201612106 [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Certo M, Moore VDG, Nishino M, Wei G, Korsmeyer S, Armstrong SA, Letai A, 2006. Mitochondria primed by death signals determine cellular addiction to antiapoptotic BCL-2 family members. Cancer Cell 9, 351–365. 10.1016/j.ccr.2006.03.027 [DOI] [PubMed] [Google Scholar]
  15. Chan DC, 2019. Mitochondrial Dynamics and Its Involvement in Disease. Annu. Rev. Pathol. Mech. Dis 10.1146/annurev-pathmechdis-012419-032711 [DOI] [PubMed] [Google Scholar]
  16. Chan DC, 2012. Fusion and Fission: Interlinked Processes Critical for Mitochondrial Health. Annu. Rev. Genet 46, 265–287. 10.1146/annurev-genet-110410-132529 [DOI] [PubMed] [Google Scholar]
  17. Chan DC, 2007. Mitochondrial Dynamics in Disease. N. Engl. J. Med 356, 1707–1709. 10.1056/NEJMp078040 [DOI] [PubMed] [Google Scholar]
  18. Chappie JS, Acharya S, Leonard M, Schmid SL, Dyda F, 2010. G domain dimerization controls dynamin’s assembly-stimulated GTPase activity. Nature 465, 435–440. 10.1038/nature09032 [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Chen H, Chan DC, 2017. Mitochondrial Dynamics in Regulating the Unique Phenotypes of Cancer and Stem Cells. Cell Metab. 26, 39–48. 10.1016/j.cmet.2017.05.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Chen H, Detmer SA, Ewald AJ, Griffin EE, Fraser SE, Chan DC, 2003. Mitofusins Mfn1 and Mfn2 coordinately regulate mitochondrial fusion and are essential for embryonic development. J. Cell Biol 160, 189–200. 10.1083/jcb.200211046 [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Chen Y, Aon MA, Hsu Y-T, Soane L, Teng X, McCaffery JM, Cheng W-C, Qi B, Li H, Alavian KN, Dayhoff-Brannigan M, Zou S, Pineda FJ, O’Rourke B, Ko YH, Pedersen PL, Kaczmarek LK, Jonas EA, Hardwick JM, 2011. Bcl-xL regulates mitochondrial energetics by stabilizing the inner membrane potential. J. Cell Biol 195, 263–276. 10.1083/jcb.201108059 [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Chipuk JE, Moldoveanu T, Llambi F, Parsons MJ, Green DR, 2010. The BCL-2 Family Reunion. Mol. Cell 37, 299–310. 10.1016/j.molcel.2010.01.025 [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Chung S, Arrell DK, Faustino RS, Terzic A, Dzeja PP, 2010. Glycolytic network restructuring integral to the energetics of embryonic stem cell cardiac differentiation. J. Mol. Cell. Cardiol. 48, 725–734. 10.1016/j.yjmcc.2009.12.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Davies VJ, Hollins AJ, Piechota MJ, Yip W, Davies JR, White KE, Nicols PP, Boulton ME, Votruba M, 2007. Opa1 deficiency in a mouse model of autosomal dominant optic atrophy impairs mitochondrial morphology, optic nerve structure and visual function. Hum. Mol. Genet 16, 1307–1318. 10.1093/hmg/ddm079 [DOI] [PubMed] [Google Scholar]
  25. Del Dotto V, Fogazza M, Carelli V, Rugolo M, Zanna C, 2018. Eight human OPA1 isoforms, long and short: What are they for? Biochim. Biophys. Acta BBA - Bioenerg 1859, 263–269. 10.1016/j.bbabio.2018.01.005 [DOI] [PubMed] [Google Scholar]
  26. Del Dotto V, Mishra P, Vidoni S, Fogazza M, Maresca A, Caporali L, McCaffery JM, Cappelletti M, Baruffini E, Lenaers G, Chan D, Rugolo M, Carelli V, Zanna C, 2017. OPA1 Isoforms in the Hierarchical Organization of Mitochondrial Functions. Cell Rep. 19, 2557–2571. 10.1016/j.celrep.2017.05.073 [DOI] [PubMed] [Google Scholar]
  27. Delettre C, Lenaers G, Griffoin J-M, Gigarel N, Lorenzo C, Belenguer P, Pelloquin L, Grosgeorge J, Turc-Carel C, Perret E, Astarie-Dequeker C, Lasquellec L, Arnaud B, Ducommun B, Kaplan J, Hamel CP, 2000. Nuclear gene OPA1, encoding a mitochondrial dynamin-related protein, is mutated in dominant optic atrophy. Nat. Genet 26, 207–210. 10.1038/79936 [DOI] [PubMed] [Google Scholar]
  28. Dhingra R, Margulets V, Chowdhury SR, Thliveris J, Jassal D, Fernyhough P, Dorn GW, Kirshenbaum LA, 2014. Bnip3 mediates doxorubicin-induced cardiac myocyte necrosis and mortality through changes in mitochondrial signaling. Proc. Natl. Acad. Sci 111, E5537–E5544. 10.1073/pnas.1414665111 [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Dix MM, Simon GM, Cravatt BF, 2008. Global mapping of the topography and magnitude of proteolytic events in apoptosis. Cell 134, 679–691. 10.1016/j.cell.2008.06.038 [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Dorn GW, 2013. Mitochondrial dynamics in heart disease. Biochim. Biophys. Acta BBA - Mol. Cell Res 1833, 233–241. 10.1016/j.bbamcr.2012.03.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Dumitru R, Gama V, Fagan BM, Bower JJ, Swahari V, Pevny LH, Deshmukh M, 2012. Human Embryonic Stem Cells Have Constitutively Active Bax at the Golgi and Are Primed to Undergo Rapid Apoptosis. Mol. Cell 46, 573–583. 10.1016/j.molcel.2012.04.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Elgass KD, Smith EA, LeGros MA, Larabell CA, Ryan MT, 2015. Analysis of ER–mitochondria contacts using correlative fluorescence microscopy and soft X-ray tomography of mammalian cells. J. Cell Sci 128, 2795–2804. 10.1242/jcs.169136 [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Elmore S, 2007. Apoptosis: A Review of Programmed Cell Death. Toxicol. Pathol 35, 495–516. 10.1080/01926230701320337 [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Escobar-Henriques M, Joaquim M, 2019. Mitofusins: Disease Gatekeepers and Hubs in Mitochondrial Quality Control by E3 Ligases. Front. Physiol 10 10.3389/fphys.2019.00517 [DOI] [PMC free article] [PubMed] [Google Scholar]
  35. Eskes R, Desagher S, Antonsson B, Martinou JC, 2000. Bid induces the oligomerization and insertion of Bax into the outer mitochondrial membrane. Mol. Cell. Biol 20, 929–935. 10.1128/mcb.20.3.929-935.2000 [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Estaquier J, Arnoult D, 2007. Inhibiting Drp1-mediated mitochondrial fission selectively prevents the release of cytochrome c during apoptosis. Cell Death Differ. 14, 1086–1094. 10.1038/sj.cdd.4402107 [DOI] [PubMed] [Google Scholar]
  37. Evans CS, Holzbaur ELF, 2019. Quality Control in Neurons: Mitophagy and Other Selective Autophagy Mechanisms. J. Mol. Biol 10.1016/j.jmb.2019.06.031 [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Evans MJ, Kaufman MH, 1981. Establishment in culture of pluripotential cells from mouse embryos. Nature 292, 154–156. 10.1038/292154a0 [DOI] [PubMed] [Google Scholar]
  39. Facucho-Oliveira JM, St. John JC, 2009. The Relationship Between Pluripotency and Mitochondrial DNA Proliferation During Early Embryo Development and Embryonic Stem Cell Differentiation. Stem Cell Rev. Rep 5, 140–158. 10.1007/s12015-009-9058-0 [DOI] [PubMed] [Google Scholar]
  40. Fernández ÁF, Sebti S, Wei Y, Zou Z, Shi M, McMillan KL, He C, Ting T, Liu Y, Chiang W-C, Marciano DK, Schiattarella GG, Bhagat G, Moe OW, Hu MC, Levine B, 2018. Disruption of the beclin 1–BCL2 autophagy regulatory complex promotes longevity in mice. Nature 558, 136–140. 10.1038/s41586-018-0162-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Fonseca TB, Sánchez-Guerrero Á, Milosevic I, Raimundo N, 2019. Mitochondrial fission requires DRP1 but not dynamins. Nature 570, E34–E42. 10.1038/s41586-019-1296-y [DOI] [PubMed] [Google Scholar]
  42. Francy CA, Clinton RW, Fröhlich C, Murphy C, Mears JA, 2017. Cryo-EM Studies of Drp1 Reveal Cardiolipin Interactions that Activate the Helical Oligomer. Sci. Rep 7, 1–12. 10.1038/s41598-017-11008-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Frank S, Gaume B, Bergmann-Leitner ES, Leitner WW, Robert EG, Catez F, Smith CL, Youle RJ, 2001. The Role of Dynamin-Related Protein 1, a Mediator of Mitochondrial Fission, in Apoptosis. Dev. Cell 1, 515–525. 10.1016/S1534-5807(01)00055-7 [DOI] [PubMed] [Google Scholar]
  44. 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, 2006. OPA1 controls apoptotic cristae remodeling independently from mitochondrial fusion. Cell 126, 177–189. 10.1016/j.cell.2006.06.025 [DOI] [PubMed] [Google Scholar]
  45. Friedman JR, Nunnari J, 2014. Mitochondrial form and function. Nature 505, 335–343. 10.1038/nature12985 [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Fu W, Liu Y, Yin H, 2019. Mitochondrial Dynamics: Biogenesis, Fission, Fusion, and Mitophagy in the Regulation of Stem Cell Behaviors. Stem Cells Int. 2019. 10.1155/2019/9757201 [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Fuchs Y, Steller H, 2011. Programmed Cell Death in Animal Development and Disease. Cell 147, 742–758. 10.1016/j.cell.2011.10.033 [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Fujita J, Crane AM, Souza MK, Dejosez M, Kyba M, Flavell RA, Thomson JA, Zwaka TP, 2008. Caspase Activity Mediates the Differentiation of Embryonic Stem Cells. Cell Stem Cell 2, 595–601. 10.1016/j.stem.2008.04.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Galluzzi L, Vitale I, Aaronson SA, Abrams JM, Adam D, Agostinis P, Alnemri ES, Altucci L, Amelio I, Andrews DW, Annicchiarico-Petruzzelli M, Antonov AV, Arama E, Baehrecke EH, Barlev NA, Bazan NG, Bernassola F, Bertrand MJM, Bianchi K, Blagosklonny MV, Blomgren K, Borner C, Boya P, Brenner C, Campanella M, Candi E, Carmona-Gutierrez D, Cecconi F, Chan FK-M, Chandel NS, Cheng EH, Chipuk JE, Cidlowski JA, Ciechanover A, Cohen GM, Conrad M, Cubillos-Ruiz JR, Czabotar PE, D’Angiolella V, Dawson TM, Dawson VL, Laurenzi VD, Maria RD, Debatin K-M, DeBerardinis RJ, Deshmukh M, Daniele ND, Virgilio FD, Dixit VM, Dixon SJ, Duckett CS, Dynlacht BD, El-Deiry WS, Elrod JW, Fimia GM, Fulda S, García-Sáez AJ, Garg AD, Garrido C, Gavathiotis E, Golstein P, Gottlieb E, Green DR, Greene LA, Gronemeyer H, Gross A, Hajnoczky G, Hardwick JM, Harris IS, Hengartner MO, Hetz C, Ichijo H, Jäättelä M, Joseph B, Jost PJ, Juin PP, Kaiser WJ, Karin M, Kaufmann T, Kepp O, Kimchi A, Kitsis RN, Klionsky DJ, Knight RA, Kumar S, Lee SW, Lemasters JJ, Levine B, Linkermann A, Lipton SA, Lockshin RA, López-Otín C, Lowe SW, Luedde T, Lugli E, MacFarlane M, Madeo F, Malewicz M, Malorni W, Manic G, Marine J-C, Martin SJ, Martinou J-C, Medema JP, Mehlen P, Meier P, Melino S, Miao EA, Molkentin JD, Moll UM, Muñoz-Pinedo C, Nagata S, Nuñez G, Oberst A, Oren M, Overholtzer M, Pagano M, Panaretakis T, Pasparakis M, Penninger JM, Pereira DM, Pervaiz S, Peter ME, Piacentini M, Pinton P, Prehn JHM, Puthalakath H, Rabinovich GA, Rehm M, Rizzuto R, Rodrigues CMP, Rubinsztein DC, Rudel T, Ryan KM, Sayan E, Scorrano L, Shao F, Shi Y, Silke J, Simon H-U, Sistigu A, Stockwell BR, Strasser A, Szabadkai G, Tait SWG, Tang D, Tavernarakis N, Thorburn A, Tsujimoto Y, Turk B, Berghe TV, Vandenabeele P, Heiden MGV, Villunger A, Virgin HW, Vousden KH, Vucic D, Wagner EF, Walczak H, Wallach D, Wang Y, Wells JA, Wood W, Yuan J, Zakeri Z, Zhivotovsky B, Zitvogel L, Melino G, Kroemer G, 2018. Molecular mechanisms of cell death: recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ. 25, 486–541. 10.1038/s41418-017-0012-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Gama V, Deshmukh M, 2012. Human embryonic stem cells: living on the edge. Cell Cycle Georget. Tex 11, 3905–3906. 10.4161/cc.22233 [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Ganesan V, Willis SD, Chang K-T, Beluch S, Cooper KF, Strich R, 2019. Cyclin C directly stimulates Drp1 GTP affinity to mediate stress-induced mitochondrial hyperfission. Mol. Biol. Cell 30, 302–311. 10.1091/mbc.E18-07-0463 [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Gao S, Malsburg A. von der, Paeschke S, Behlke J, Haller O, Kochs G, Daumke O, 2010. Structural basis of oligomerization in the stalk region of dynamin-like MxA. Nature 465, 502–506. 10.1038/nature08972 [DOI] [PubMed] [Google Scholar]
  53. Gao W, Pu Y, Luo KQ, Chang DC, 2001. Temporal relationship between cytochrome c release and mitochondrial swelling during UV-induced apoptosis in living HeLa cells. J. Cell Sci 114, 2855–2862. [DOI] [PubMed] [Google Scholar]
  54. Garrido C, Galluzzi L, Brunet M, Puig PE, Didelot C, Kroemer G, 2006. Mechanisms of cytochrome c release from mitochondria. Cell Death Differ. 13, 1423–1433. 10.1038/sj.cdd.4401950 [DOI] [PubMed] [Google Scholar]
  55. Gavathiotis E, Suzuki M, Davis ML, Pitter K, Bird GH, Katz SG, Tu H-C, Kim H, Cheng EH-Y, Tjandra N, Walensky LD, 2008. BAX Activation is Initiated at a Novel Interaction Site. Nature 455, 1076–1081. 10.1038/nature07396 [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Gegg ME, Cooper JM, Chau K-Y, Rojo M, Schapira AHV, Taanman J-W, 2010. Mitofusin 1 and mitofusin 2 are ubiquitinated in a PINK1/parkin-dependent manner upon induction of mitophagy. Hum. Mol. Genet 19, 4861–4870. 10.1093/hmg/ddq419 [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Goldstein JC, Waterhouse NJ, Juin P, Evan GI, Green DR, 2000. The coordinate release of cytochrome c during apoptosis is rapid, complete and kinetically invariant. Nat. Cell Biol 2, 156–162. 10.1038/35004029 [DOI] [PubMed] [Google Scholar]
  58. Goodell MA, Nguyen H, Shroyer N, 2015. Somatic stem cell heterogeneity: diversity in the blood, skin and intestinal stem cell compartments. Nat. Rev. Mol. Cell Biol 16, 299–309. 10.1038/nrm3980 [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Goping IS, Gross A, Lavoie JN, Nguyen M, Jemmerson R, Roth K, Korsmeyer SJ, Shore GC, 1998. Regulated Targeting of BAX to Mitochondria. J. Cell Biol 143, 207–215. 10.1083/jcb.143.1.207 [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Green DR, Chipuk JE, 2008. APOPTOSIS: Stabbed in the BAX. Nature 455, 1047–1049. 10.1038/4551047a [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Hardwick JM, Soane L, 2013. Multiple Functions of BCL-2 Family Proteins. Cold Spring Harb. Perspect. Biol 5 10.1101/cshperspect.a008722 [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Hinson JT, Chopra A, Nafissi N, Polacheck WJ, Benson CC, Swist S, Gorham J, Yang L, Schafer S, Sheng CC, Haghighi A, Homsy J, Hubner N, Church G, Cook SA, Linke WA, Chen CS, Seidman JG, Seidman CE, 2015. Titin mutations in iPS cells define sarcomere insufficiency as a cause of dilated cardiomyopathy. Science 349, 982–986. 10.1126/science.aaa5458 [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Hollville E, Carroll RG, Cullen SP, Martin SJ, 2014. Bcl-2 Family Proteins Participate in Mitochondrial Quality Control by Regulating Parkin/PINK1-Dependent Mitophagy. Mol. Cell 55, 451–466. 10.1016/j.molcel.2014.06.001 [DOI] [PubMed] [Google Scholar]
  64. Hoppins S, Edlich F, Cleland MM, Banerjee S, McCaffery JM, Youle RJ, Nunnari J, 2011. The Soluble Form of Bax Regulates Mitochondrial Fusion via MFN2 Homotypic Complexes. Mol. Cell 41, 150–160. 10.1016/j.molcel.2010.11.030 [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Hoppins S, Lackner L, Nunnari J, 2007. The Machines that Divide and Fuse Mitochondria. Annu. Rev. Biochem 76, 751–780. 10.1146/annurev.biochem.76.071905.090048 [DOI] [PubMed] [Google Scholar]
  66. Hsu Y-T, Youle RJ, 1997. Nonionic Detergents Induce Dimerization among Members of the Bcl-2 Family. J. Biol. Chem 272, 13829–13834. 10.1074/jbc.272.21.13829 [DOI] [PubMed] [Google Scholar]
  67. Ingerman E, Perkins EM, Marino M, Mears JA, McCaffery JM, Hinshaw JE, Nunnari J, 2005. Dnm1 forms spirals that are structurally tailored to fit mitochondria. J. Cell Biol 170, 1021–1027. 10.1083/jcb.200506078 [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Inoue S, Browne G, Melino G, Cohen GM, 2009. Ordering of caspases in cells undergoing apoptosis by the intrinsic pathway. Cell Death Differ. 16, 1053–1061. 10.1038/cdd.2009.29 [DOI] [PubMed] [Google Scholar]
  69. Ishihara N, Nomura M, Jofuku A, Kato H, Suzuki SO, Masuda K, Otera H, Nakanishi Y, Nonaka I, Goto Y, Taguchi N, Morinaga H, Maeda M, Takayanagi R, Yokota S, Mihara K, 2009. Mitochondrial fission factor Drp1 is essential for embryonic development and synapse formation in mice. Nat. Cell Biol 11, 958–966. 10.1038/ncb1907 [DOI] [PubMed] [Google Scholar]
  70. Jahani-Asl A, Germain M, Slack RS, 2010. Mitochondria: Joining forces to thwart cell death. Biochim. Biophys. Acta BBA - Mol. Basis Dis., Mitochondrial Dysfunction 1802, 162–166. 10.1016/j.bbadis.2009.09.006 [DOI] [PubMed] [Google Scholar]
  71. Jain AK, Barton MC, 2018. p53: emerging roles in stem cells, development and beyond. Development 145, dev158360 10.1242/dev.158360 [DOI] [PubMed] [Google Scholar]
  72. Jiang X, Jiang H, Shen Z, Wang X, 2014. Activation of mitochondrial protease OMA1 by Bax and Bak promotes cytochrome c release during apoptosis. Proc. Natl. Acad. Sci 111, 14782–14787. 10.1073/pnas.1417253111 [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Johnson LV, Walsh ML, Bockus BJ, Chen LB, 1981. Monitoring of relative mitochondrial membrane potential in living cells by fluorescence microscopy. J. Cell Biol 88, 526–535. 10.1083/jcb.88.3.526 [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Kageyama Y, Hoshijima M, Seo K, Bedja D, Sysa-Shah P, Andrabi SA, Chen W, Höke A, Dawson VL, Dawson TM, Gabrielson KL, Kass DA, Iijima M, Sesaki H, 2014. Parkin-independent mitophagy requires Drp1 and maintains the integrity of mammalian heart and brain. EMBO J. 33, 2798–2813. 10.15252/embj.201488658 [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Kale J, Osterlund EJ, Andrews DW, 2018. BCL-2 family proteins: changing partners in the dance towards death. Cell Death Differ. 25, 65–80. 10.1038/cdd.2017.186 [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Kalkavan H, Green DR, 2018. MOMP, cell suicide as a BCL-2 family business. Cell Death Differ. 25, 46–55. 10.1038/cdd.2017.179 [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Kamerkar SC, Kraus F, Sharpe AJ, Pucadyil TJ, Ryan MT, 2018. Dynamin-related protein 1 has membrane constricting and severing abilities sufficient for mitochondrial and peroxisomal fission. Nat. Commun. 9, 5239 10.1038/s41467-018-07543-w [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Karbowski M, Lee Y-J, Gaume B, Jeong S-Y, Frank S, Nechushtan A, Santel A, Fuller M, Smith CL, Youle RJ, 2002. Spatial and temporal association of Bax with mitochondrial fission sites, Drp1, and Mfn2 during apoptosis. J. Cell Biol. 159, 931–938. 10.1083/jcb.200209124 [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Karbowski M, Norris KL, Cleland MM, Jeong S-Y, Youle RJ, 2006. Role of Bax and Bak in mitochondrial morphogenesis. Nature 443, 658–662. 10.1038/nature05111 [DOI] [PubMed] [Google Scholar]
  80. Kawamura T, Suzuki J, Wang YV, Menendez S, Morera LB, Raya A, Wahl GM, Belmonte JCI, 2009. Linking the p53 tumour suppressor pathway to somatic cell reprogramming. Nature 460, 1140–1144. 10.1038/nature08311 [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Kerr JFR, Wyllie AH, Currie AR, 1972. Apoptosis: A Basic Biological Phenomenon with Wideranging Implications in Tissue Kinetics. Br. J. Cancer 26, 239–257. 10.1038/bjc.1972.33 [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Kim H-E, Du F, Fang M, Wang X, 2005. Formation of apoptosome is initiated by cytochrome c-induced dATP hydrolysis and subsequent nucleotide exchange on Apaf-1. Proc. Natl. Acad. Sci 102, 17545–17550. 10.1073/pnas.0507900102 [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Kornmann B, Walter P, 2010. ERMES-mediated ER-mitochondria contacts: molecular hubs for the regulation of mitochondrial biology. J. Cell Sci 123, 1389–1393. 10.1242/jcs.058636 [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Korobova F, Ramabhadran V, Higgs HN, 2013. An actin-dependent step in mitochondrial fission mediated by the ER-associated formin INF2. Science 339, 464–467. 10.1126/science.1228360 [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Kruse J-P, Gu W, 2009. Modes of p53 Regulation. Cell 137, 609–622. 10.1016/j.cell.2009.04.050 [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Kuznetsov AV, Hermann M, Saks V, Hengster P, Margreiter R, 2009. The cell-type specificity of mitochondrial dynamics. Int. J. Biochem. Cell Biol., Mitochondrial Dynamics and Function in Biology and Medicine 41, 1928–1939. 10.1016/j.biocel.2009.03.007 [DOI] [PubMed] [Google Scholar]
  87. Leber B, Lin J, Andrews DW, 2007. Embedded Together: The Life and Death Consequences of Interaction of the Bcl-2 Family with Membranes. Apoptosis Int. J. Program. Cell Death 12, 897–911. 10.1007/s10495-007-0746-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Lee JE, Westrate LM, Wu H, Page C, Voeltz GK, 2016. Multiple Dynamin family members collaborate to drive mitochondrial division. Nature 540, 139–143. 10.1038/nature20555 [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Lee Y, Jeong S-Y, Karbowski M, Smith CL, Youle RJ, 2004. Roles of the Mammalian Mitochondrial Fission and Fusion Mediators Fis1, Drp1, and Opa1 in Apoptosis. Mol. Biol. Cell 15, 5001–5011. 10.1091/mbc.e04-04-0294 [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Legros F, Lombès A, Frachon P, Rojo M, 2002. Mitochondrial Fusion in Human Cells Is Efficient, Requires the Inner Membrane Potential, and Is Mediated by Mitofusins. Mol. Biol. Cell 13, 4343–4354. 10.1091/mbc.e02-06-0330 [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Levine AJ, Puzio-Kuter AM, Chan CS, Hainaut P, 2016. The Role of the p53 Protein in Stem-Cell Biology and Epigenetic Regulation. Cold Spring Harb. Perspect. Med 6, a026153 10.1101/cshperspect.a026153 [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Lewis MR, Lewis WH, 1914. Mitochondria in Tissue Culture. Science 39, 330–333. 10.1126/science.39.1000.330 [DOI] [PubMed] [Google Scholar]
  93. Lewis SC, Uchiyama LF, Nunnari J, 2016. ER-mitochondria contacts couple mtDNA synthesis with mitochondrial division in human cells. Science 353, aaf5549 10.1126/science.aaf5549 [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Li F, He Z, Shen J, Huang Q, Li W, Liu X, He Y, Wolf F, Li C-Y, 2010. Apoptotic Caspases Regulate Induction of iPSCs from Human Fibroblasts. Cell Stem Cell 7, 508–520. 10.1016/j.stem.2010.09.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Li H, Chen Y, Jones AF, Sanger RH, Collis LP, Flannery R, McNay EC, Yu T, Schwarzenbacher R, Bossy B, Bossy-Wetzel E, Bennett MVL, Pypaert M, Hickman JA, Smith PJS, Hardwick JM, Jonas EA, 2008. Bcl-xL induces Drp1-dependent synapse formation in cultured hippocampal neurons. Proc. Natl. Acad. Sci 105, 2169–2174. 10.1073/pnas.0711647105 [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Li P, Nijhawan D, Budihardjo I, Srinivasula SM, Ahmad M, Alnemri ES, Wang X, 1997. Cytochrome c and dATP-dependent formation of Apaf-1/caspase-9 complex initiates an apoptotic protease cascade. Cell 91, 479–489. 10.1016/s0092-8674(00)80434-1 [DOI] [PubMed] [Google Scholar]
  97. Li Y, Feng H, Gu H, Lewis DW, Yuan Y, Zhang L, Yu H, Zhang P, Cheng H, Miao W, Yuan W, Cheng S-Y, Gollin SM, Cheng T, 2013. The p53–PUMA axis suppresses iPSC generation. Nat. Commun 4, 1–9. 10.1038/ncomms3174 [DOI] [PMC free article] [PubMed] [Google Scholar]
  98. Lindsten T, Ross AJ, King A, Zong WX, Rathmell JC, Shiels HA, Ulrich E, Waymire KG, Mahar P, Frauwirth K, Chen Y, Wei M, Eng VM, Adelman DM, Simon MC, Ma A, Golden JA, Evan G, Korsmeyer SJ, MacGregor GR, Thompson CB, 2000. The combined functions of proapoptotic Bcl-2 family members bak and bax are essential for normal development of multiple tissues. Mol. Cell 6, 1389–1399. 10.1016/s1097-2765(00)00136-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Liu JC, Guan X, Ryan JA, Rivera AG, Mock C, Agarwal V, Letai A, Lerou PH, Lahav G, 2013. High Mitochondrial Priming Sensitizes hESCs to DNA-Damage-Induced Apoptosis. Cell Stem Cell 13, 483–491. 10.1016/j.stem.2013.07.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Liu X, Kim CN, Yang J, Jemmerson R, Wang X, 1996. Induction of Apoptotic Program in Cell-Free Extracts: Requirement for dATP and Cytochrome c. Cell 86, 147–157. 10.1016/S0092-8674(00)80085-9 [DOI] [PubMed] [Google Scholar]
  101. Llambi F, Moldoveanu T, Tait SWG, Bouchier-Hayes L, Temirov J, McCormick LL, Dillon CP, Green DR, 2011. A unified model of mammalian BCL-2 protein family interactions at the mitochondria. Mol. Cell 44, 517–531. 10.1016/j.molcel.2011.10.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Llambi F, Wang Y-M, Victor B, Yang M, Schneider DM, Gingras S, Parsons MJ, Zheng JH, Brown SA, Pelletier S, Moldoveanu T, Chen T, Green DR, 2016. BOK Is a Non-Canonical BCL-2 Family Effector of Apoptosis Regulated by ER-Associated Degradation. Cell 165, 421–433. 10.1016/j.cell.2016.02.026 [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. MacVicar T, Langer T, 2016. OPA1 processing in cell death and disease – the long and short of it. J. Cell Sci. 129, 2297–2306. 10.1242/jcs.159186 [DOI] [PubMed] [Google Scholar]
  104. Madden DT, Davila-Kruger D, Melov S, Bredesen DE, 2011. Human Embryonic Stem Cells Express Elevated Levels of Multiple Pro-Apoptotic BCL-2 Family Members. PLoS ONE 6 10.1371/journal.pone.0028530 [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Mahrus S, Trinidad JC, Barkan DT, Sali A, Burlingame AL, Wells JA, 2008. Global sequencing of proteolytic cleavage sites in apoptosis by specific labeling of protein N termini. Cell 134, 866–876. 10.1016/j.cell.2008.08.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Maiuri MC, Le Toumelin G, Criollo A, Rain J-C, Gautier F, Juin P, Tasdemir E, Pierron G, Troulinaki K, Tavernarakis N, Hickman JA, Geneste O, Kroemer G, 2007. Functional and physical interaction between Bcl-XL and a BH3-like domain in Beclin-1. EMBO J. 26, 2527–2539. 10.1038/sj.emboj.7601689 [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Martin GR, 1981. Isolation of a pluripotent cell line from early mouse embryos cultured in medium conditioned by teratocarcinoma stem cells. Proc. Natl. Acad. Sci. U. S. A. 78, 7634–7638. [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Mears JA, Lackner LL, Fang S, Ingerman E, Nunnari J, Hinshaw JE, 2011. Conformational changes in Dnm1 support a contractile mechanism for mitochondrial fission. Nat. Struct. Mol. Biol. 18, 20–26. 10.1038/nsmb.1949 [DOI] [PMC free article] [PubMed] [Google Scholar]
  109. Meeusen S, McCaffery JM, Nunnari J, 2004. Mitochondrial Fusion Intermediates Revealed in Vitro. Science 305, 1747–1752. 10.1126/science.1100612 [DOI] [PubMed] [Google Scholar]
  110. Meglei G, McQuibban GA, 2009. The Dynamin-Related Protein Mgm1p Assembles into Oligomers and Hydrolyzes GTP To Function in Mitochondrial Membrane Fusion. Biochemistry 48, 1774–1784. 10.1021/bi801723d [DOI] [PubMed] [Google Scholar]
  111. Mehlen P, Bredesen DE, 2011. Dependence Receptors: From Basic Research to Drug Development. Sci. Signal 4, mr2–mr2. 10.1126/scisignal.2001521 [DOI] [PubMed] [Google Scholar]
  112. Mishra P, Carelli V, Manfredi G, Chan DC, 2014. Proteolytic Cleavage of Opa1 Stimulates Mitochondrial Inner Membrane Fusion and Couples Fusion to Oxidative Phosphorylation. Cell Metab. 19, 630–641. 10.1016/j.cmet.2014.03.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Moldoveanu T, Czabotar PE, 2019. BAX, BAK, and BOK: A Coming of Age for the BCL-2 Family Effector Proteins. Cold Spring Harb. Perspect. Biol a036319 10.1101/cshperspect.a036319 [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Montero J, Letai A, 2016. Dynamic BH3 profiling-poking cancer cells with a stick. Mol. Cell. Oncol 3, e1040144 10.1080/23723556.2015.1040144 [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Montero J, Sarosiek KA, DeAngelo JD, Maertens O, Ryan J, Ercan D, Piao H, Horowitz NS, Berkowitz RS, Matulonis U, Jänne PA, Amrein PC, Cichowski K, Drapkin R, Letai A, 2015. Drug-Induced Death Signaling Strategy Rapidly Predicts Cancer Response to Chemotherapy. Cell 160, 977–989. 10.1016/j.cell.2015.01.042 [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Montessuit S, Somasekharan SP, Terrones O, Lucken-Ardjomande S, Herzig S, Schwarzenbacher R, Manstein DJ, Bossy-Wetzel E, Basañez G, Meda P, Martinou J-C, 2010. Membrane Remodeling Induced by the Dynamin-Related Protein Drp1 Stimulates Bax Oligomerization. Cell 142, 889–901. 10.1016/j.cell.2010.08.017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Morciano G, Giorgi C, Balestra D, Marchi S, Perrone D, Pinotti M, Pinton P, 2016a. Mcl-1 involvement in mitochondrial dynamics is associated with apoptotic cell death. Mol. Biol. Cell 27, 20–34. 10.1091/mbc.E15-01-0028 [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Morciano G, Pedriali G, Sbano L, Iannitti T, Giorgi C, Pinton P, 2016b. Intersection of mitochondrial fission and fusion machinery with apoptotic pathways: Role of Mcl-1: Mcl-1 in mitochondrial dynamics. Biol. Cell 108, 279–293. 10.1111/boc.201600019 [DOI] [PubMed] [Google Scholar]
  119. Naik PP, Birbrair A, Bhutia SK, 2019. Mitophagy-driven metabolic switch reprograms stem cell fate. Cell. Mol. Life Sci 76, 27–43. 10.1007/s00018-018-2922-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Nakamura A, Swahari V, Plestant C, Smith I, McCoy E, Smith S, Moy SS, Anton ES, Deshmukh M, 2016. Bcl-xL Is Essential for the Survival and Function of Differentiated Neurons in the Cortex That Control Complex Behaviors. J. Neurosci. Off. J. Soc. Neurosci 36, 5448–5461. 10.1523/JNEUROSCI.4247-152016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  121. Narendra D, Tanaka A, Suen D-F, Youle RJ, 2008. Parkin is recruited selectively to impaired mitochondria and promotes their autophagy. J. Cell Biol 183, 795–803. 10.1083/jcb.200809125 [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Nunnari J, Marshall WF, Straight A, Murray A, Sedat JW, Walter P, 1997. Mitochondrial transmission during mating in Saccharomyces cerevisiae is determined by mitochondrial fusion and fission and the intramitochondrial segregation of mitochondrial DNA. Mol. Biol. Cell 8, 1233–1242. 10.1091/mbc.8.7.1233 [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Oltval ZN, Milliman CL, Korsmeyer SJ, 1993. Bcl-2 heterodimerizes in vivo with a conserved homolog, Bax, that accelerates programed cell death. Cell 74, 609–619. 10.1016/0092-8674(93)90509-O [DOI] [PubMed] [Google Scholar]
  124. Onoue K, Jofuku A, Ban-Ishihara R, Ishihara T, Maeda M, Koshiba T, Itoh T, Fukuda M, Otera H, Oka T, Takano H, Mizushima N, Mihara K, Ishihara N, 2013. Fis1 acts as a mitochondrial recruitment factor for TBC1D15 that is involved in regulation of mitochondrial morphology. J. Cell Sci 126, 176–185. 10.1242/jcs.111211 [DOI] [PubMed] [Google Scholar]
  125. Ow Y-LP, Green DR, Hao Z, Mak TW, 2008. Cytochrome c : functions beyond respiration. Nat. Rev. Mol. Cell Biol 9, 532–542. 10.1038/nrm2434 [DOI] [PubMed] [Google Scholar]
  126. Pattingre S, Tassa A, Qu X, Garuti R, Liang XH, Mizushima N, Packer M, Schneider MD, Levine B, 2005. Bcl-2 Antiapoptotic Proteins Inhibit Beclin 1-Dependent Autophagy. Cell 122, 927–939. 10.1016/j.cell.2005.07.002 [DOI] [PubMed] [Google Scholar]
  127. Pickles S, Vigié P, Youle RJ, 2018. Mitophagy and Quality Control Mechanisms in Mitochondrial Maintenance. Curr. Biol 28, R170–R185. 10.1016/j.cub.2018.01.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. Pon LA, 2013. Mitochondrial Fission: Rings around the Organelle. Curr. Biol 23, R279–R281. 10.1016/j.cub.2013.02.042 [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. Praefcke GJK, McMahon HT, 2004. The dynamin superfamily: universal membrane tubulation and fission molecules? Nat. Rev. Mol. Cell Biol 5, 133–147. 10.1038/nrm1313 [DOI] [PubMed] [Google Scholar]
  130. Prieto J, León M, Ponsoda X, Sendra R, Bort R, Ferrer-Lorente R, Raya A, López-García C, Torres J, 2016. Early ERK1/2 activation promotes DRP1-dependent mitochondrial fission necessary for cell reprogramming. Nat. Commun 7, 11124 10.1038/ncomms11124 [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Prigione A, Hossini AM, Lichtner B, Serin A, Fauler B, Megges M, Lurz R, Lehrach H, Makrantonaki E, Zouboulis CC, Adjaye J, 2011. Mitochondrial-Associated Cell Death Mechanisms Are Reset to an Embryonic-Like State in Aged Donor-Derived iPS Cells Harboring Chromosomal Aberrations. PLoS ONE 6 10.1371/journal.pone.0027352 [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Purvis JE, Karhohs KW, Mock C, Batchelor E, Loewer A, Lahav G, 2012. p53 Dynamics Control Cell Fate. Science 336, 1440–1444. 10.1126/science.1218351 [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Ramirez MLG, Salvesen GS, 2018. A primer on caspase mechanisms. Semin. Cell Dev. Biol., SI: Nuclear positioning 82, 79–85. 10.1016/j.semcdb.2018.01.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. Rasmussen ML, Kline LA, Park KP, Ortolano NA, Romero-Morales AI, Anthony CC, Beckermann KE, Gama V, 2018. A Non-apoptotic Function of MCL-1 in Promoting Pluripotency and Modulating Mitochondrial Dynamics in Stem Cells. Stem Cell Rep. 10, 684–692. 10.1016/j.stemcr.2018.01.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. Rastogi A, Joshi P, Contreras E, Gama V, 2019. Remodeling of mitochondrial morphology and function: an emerging hallmark of cellular reprogramming. Cell Stress 3, 181–194. 10.15698/cst2019.06.189 [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Rehklau K, Hoffmann L, Gurniak CB, Ott M, Witke W, Scorrano L, Culmsee C, Rust MB, 2017. Cofilin1-dependent actin dynamics control DRP1-mediated mitochondrial fission. Cell Death Dis. 8, e3063 10.1038/cddis.2017.448 [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Rinkenberger JL, Horning S, Klocke B, Roth K, Korsmeyer SJ, 2000. Mcl-1 deficiency results in peri-implantation embryonic lethality. Genes Dev. 6. [PMC free article] [PubMed] [Google Scholar]
  138. Rizzuto R, Brini M, Giorgi FD, Rossi R, Heim R, Tsien RY, Pozzan T, 1996. Double labelling of subcellular structures with organelle-targeted GFP mutants in vivo. Curr. Biol. 6, 183–188. 10.1016/S0960-9822(02)00451-7 [DOI] [PubMed] [Google Scholar]
  139. Salisbury-Ruf CT, Bertram CC, Vergeade A, Lark DS, Shi Q, Heberling ML, Fortune NL, Okoye GD, Jerome WG, Wells QS, Fessel J, Moslehi J, Chen H, Roberts LJ, Boutaud O, Gamazon ER, Zinkel SS, 2018. Bid maintains mitochondrial cristae structure and function and protects against cardiac disease in an integrative genomics study. eLife 7, e40907 10.7554/eLife.40907 [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Santel A, Frank S, Gaume B, Herrler M, Youle RJ, Fuller MT, 2003. Mitofusin-1 protein is a generally expressed mediator of mitochondrial fusion in mammalian cells. J. Cell Sci 116, 2763–2774. 10.1242/jcs.00479 [DOI] [PubMed] [Google Scholar]
  141. Schrepfer E, Scorrano L, 2016. Mitofusins, from Mitochondria to Metabolism. Mol. Cell 61, 683–694. 10.1016/j.molcel.2016.02.022 [DOI] [PubMed] [Google Scholar]
  142. Schulman JJ, Szczesniak LM, Bunker EN, Nelson HA, Roe MW, Wagner LE, Yule DI, Wojcikiewicz RJH, 2019. Bok regulates mitochondrial fusion and morphology. Cell Death Differ. 26, 2682–2694. 10.1038/s41418-019-0327-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  143. Schulman JJ, Wright FA, Han X, Zluhan EJ, Szczesniak LM, Wojcikiewicz RJH, 2016. The Stability and Expression Level of Bok Are Governed by Binding to Inositol 1,4,5-Trisphosphate Receptors. J. Biol. Chem 291, 11820–11828. 10.1074/jbc.M115.711242 [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. Shamas-Din A, Kale J, Leber B, Andrews DW, 2013. Mechanisms of action of Bcl-2 family proteins. Cold Spring Harb. Perspect. Biol 5, a008714 10.1101/cshperspect.a008714 [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. Sheridan C, Delivani P, Cullen SP, Martin SJ, 2008. Bax- or Bak-Induced Mitochondrial Fission Can Be Uncoupled from Cytochrome c Release. Mol. Cell 31, 570–585. 10.1016/j.molcel.2008.08.002 [DOI] [PubMed] [Google Scholar]
  146. Sinibaldi F, Howes BD, Droghetti E, Polticelli F, Piro MC, Di Pierro D, Fiorucci L, Coletta M, Smulevich G, Santucci R, 2013. Role of Lysines in Cytochrome c–Cardiolipin Interaction. Biochemistry 52, 4578–4588. 10.1021/bi400324c [DOI] [PubMed] [Google Scholar]
  147. Smirnova E, Griparic L, Shurland D-L, van der Bliek AM, 2001. Dynamin-related Protein Drp1 Is Required for Mitochondrial Division in Mammalian Cells. Mol. Biol. Cell 12, 2245–2256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  148. Smith RC, Tabar V, 2019. Constructing and Deconstructing Cancers using Human Pluripotent Stem Cells and Organoids. Cell Stem Cell 24, 12–24. 10.1016/j.stem.2018.11.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  149. Stavru F, Palmer AE, Wang C, Youle RJ, Cossart P, 2013. Atypical mitochondrial fission upon bacterial infection. Proc. Natl. Acad. Sci 110, 16003–16008. 10.1073/pnas.1315784110 [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. Steffen J, Koehler CM, 2018. ER–mitochondria contacts: Actin dynamics at the ER control mitochondrial fission via calcium release. J. Cell Biol 217, 15–17. 10.1083/jcb.201711075 [DOI] [PMC free article] [PubMed] [Google Scholar]
  151. Sugioka R, Shimizu S, Tsujimoto Y, 2004. Fzo1, a Protein Involved in Mitochondrial Fusion, Inhibits Apoptosis. J. Biol. Chem 279, 52726–52734. 10.1074/jbc.M408910200 [DOI] [PubMed] [Google Scholar]
  152. Taguchi N, Ishihara N, Jofuku A, Oka T, Mihara K, 2007. Mitotic Phosphorylation of Dynamin-related GTPase Drp1 Participates in Mitochondrial Fission. J. Biol. Chem 282, 11521–11529. 10.1074/jbc.M607279200 [DOI] [PubMed] [Google Scholar]
  153. Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, Yamanaka S, 2007. Induction of Pluripotent Stem Cells from Adult Human Fibroblasts by Defined Factors. Cell 131, 861–872. 10.1016/j.cell.2007.11.019 [DOI] [PubMed] [Google Scholar]
  154. Takahashi K, Yamanaka S, 2015. A developmental framework for induced pluripotency. Development 142, 3274–3285. 10.1242/dev.114249 [DOI] [PubMed] [Google Scholar]
  155. Takahashi K, Yamanaka S, 2006. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126, 663–676. 10.1016/j.cell.2006.07.024 [DOI] [PubMed] [Google Scholar]
  156. Tanaka A, Youle RJ, 2008. A Chemical Inhibitor of DRP1 Uncouples Mitochondrial Fission and Apoptosis. Mol. Cell 29, 409–410. 10.1016/j.molcel.2008.02.005 [DOI] [PubMed] [Google Scholar]
  157. Taylor RC, Cullen SP, Martin SJ, 2008. Apoptosis: controlled demolition at the cellular level. Nat. Rev. Mol. Cell Biol 9, 231–241. 10.1038/nrm2312 [DOI] [PubMed] [Google Scholar]
  158. Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ, Marshall VS, Jones JM, 1998. Embryonic Stem Cell Lines Derived from Human Blastocysts. Science 282, 1145–1147. 10.1126/science.282.5391.1145 [DOI] [PubMed] [Google Scholar]
  159. Todt F, Cakir Z, Reichenbach F, Emschermann F, Lauterwasser J, Kaiser A, Ichim G, Tait SW, Frank S, Langer HF, Edlich F, 2015. Differential retrotranslocation of mitochondrial Bax and Bak. EMBO J. 34, 67–80. 10.15252/embj.201488806 [DOI] [PMC free article] [PubMed] [Google Scholar]
  160. Tsujimoto Y, Finger LR, Yunis J, Nowell PC, Croce CM, 1984. Cloning of the chromosome breakpoint of neoplastic B cells with the t(14;18) chromosome translocation. Science 226, 1097–1099. 10.1126/science.6093263 [DOI] [PubMed] [Google Scholar]
  161. Upton J-P, Valentijn AJ, Zhang L, Gilmore AP, 2007. The N-terminal conformation of Bax regulates cell commitment to apoptosis. Cell Death Differ. 14, 932–942. 10.1038/sj.cdd.4402092 [DOI] [PMC free article] [PubMed] [Google Scholar]
  162. van der Bliek AM, Payne GS, 2010. Dynamin Subunit Interactions Revealed. Dev. Cell 18, 687–688. 10.1016/j.devcel.2010.05.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Vásquez-Trincado C, García-Carvajal I, Pennanen C, Parra V, Hill JA, Rothermel BA, Lavandero S, 2016. Mitochondrial dynamics, mitophagy and cardiovascular disease. J. Physiol 594, 509–525. 10.1113/JP271301 [DOI] [PMC free article] [PubMed] [Google Scholar]
  164. Vazquez-Martin A, Cufí S, Corominas-Faja B, Oliveras-Ferraros C, Vellon L, Menendez JA, 2012. Mitochondrial fusion by pharmacological manipulation impedes somatic cell reprogramming to pluripotency: New insight into the role of mitophagy in cell stemness. Aging 4, 393–401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  165. Vazquez-Martin A, Van den Haute C, Cufí S, Corominas-Faja B, Cuyàs E, Lopez-Bonet E, Rodriguez-Gallego E, Fernández-Arroyo S, Joven J, Baekelandt V, Menendez JA, 2016. Mitophagy-driven mitochondrial rejuvenation regulates stem cell fate. Aging 8, 1330–1349. 10.18632/aging.100976 [DOI] [PMC free article] [PubMed] [Google Scholar]
  166. Wanet A, Arnould T, Najimi M, Renard P, 2015. Connecting Mitochondria, Metabolism, and Stem Cell Fate. Stem Cells Dev. 24, 1957–1971. 10.1089/scd.2015.0117 [DOI] [PMC free article] [PubMed] [Google Scholar]
  167. Wang K, Yin XM, Chao DT, Milliman CL, Korsmeyer SJ, 1996. BID: a novel BH3 domain-only death agonist. Genes Dev. 10, 2859–2869. 10.1101/gad.10.22.2859 [DOI] [PubMed] [Google Scholar]
  168. Wang Ping, Wang Peiguo, Liu B, Zhao J, Pang Q, Agrawal SG, Jia L, Liu F-T, 2015. Dynamin-related protein Drp1 is required for Bax translocation to mitochondria in response to irradiation-induced apoptosis. Oncotarget 6, 22598–22612. 10.18632/oncotarget.4200 [DOI] [PMC free article] [PubMed] [Google Scholar]
  169. Waterham HR, Koster J, van Roermund CWT, Mooyer PAW, Wanders RJA, Leonard JV, 2007. A Lethal Defect of Mitochondrial and Peroxisomal Fission. N. Engl. J. Med 356, 1736–1741. 10.1056/NEJMoa064436 [DOI] [PubMed] [Google Scholar]
  170. Westermann B, 2010. Mitochondrial fusion and fission in cell life and death. Nat. Rev. Mol. Cell Biol 11, 872–884. 10.1038/nrm3013 [DOI] [PubMed] [Google Scholar]
  171. Wolter KG, Hsu Y-T, Smith CL, Nechushtan A, Xi X-G, Youle RJ, 1997. Movement of Bax from the Cytosol to Mitochondria during Apoptosis. J. Cell Biol 139, 1281–1292. 10.1083/jcb.139.5.1281 [DOI] [PMC free article] [PubMed] [Google Scholar]
  172. Yamaguchi R, Lartigue L, Perkins G, Scott RT, Dixit A, Kushnareva Y, Kuwana T, Ellisman MH, Newmeyer DD, 2008. Opa1-mediated cristae opening is Bax/Bak and BH3 dependent, required for apoptosis, and independent of Bak oligomerization. Mol. Cell 31, 557–569. 10.1016/j.molcel.2008.07.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  173. Yang C, Svitkina TM, 2019. Ultrastructure and dynamics of the actin−myosin II cytoskeleton during mitochondrial fission. Nat. Cell Biol 21, 603–613. 10.1038/s41556-019-0313-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  174. Yang T, Buchan HL, Townsend KJ, Craig RW, 1996. MCL-1, a member of the BCL-2 family, is induced rapidly in response to signals for cell differentiation or death, but not to signals for cell proliferation. J. Cell. Physiol 166, 523–536. [DOI] [PubMed] [Google Scholar]
  175. Youle RJ, Bliek A.M. van der, 2012. Mitochondrial Fission, Fusion, and Stress. Science 337, 1062–1065. 10.1126/science.1219855 [DOI] [PMC free article] [PubMed] [Google Scholar]
  176. Zhang J, 2013. Autophagy and mitophagy in cellular damage control. Redox Biol. 1, 19–23. 10.1016/j.redox.2012.11.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  177. Zhang J, Ney PA, 2009. Role of BNIP3 and NIX in cell death, autophagy, and mitophagy. Cell Death Differ. 16, 939–946. 10.1038/cdd.2009.16 [DOI] [PMC free article] [PubMed] [Google Scholar]
  178. Zheng JH, Grace CR, Guibao CD, McNamara DE, Llambi F, Wang Y-M, Chen T, Moldoveanu T, 2018. Intrinsic Instability of BOK Enables Membrane Permeabilization in Apoptosis. Cell Rep. 23, 2083–2094.e6. 10.1016/j.celrep.2018.04.060 [DOI] [PMC free article] [PubMed] [Google Scholar]
  179. Zou H, Henzel WJ, Liu X, Lutschg A, Wang X, 1997. Apaf-1, a human protein homologous to C. elegans CED-4, participates in cytochrome c-dependent activation of caspase-3. Cell 90, 405–413. 10.1016/s0092-8674(00)80501-2 [DOI] [PubMed] [Google Scholar]
  180. Züchner S, Mersiyanova IV, Muglia M, Bissar-Tadmouri N, Rochelle J, Dadali EL, Zappia M, Nelis E, Patitucci A, Senderek J, Parman Y, Evgrafov O, Jonghe PD, Takahashi Y, Tsuji S, Pericak-Vance MA, Quattrone A, Battologlu E, Polyakov AV, Timmerman V, Schröder JM, Vance JM, 2004. Mutations in the mitochondrial GTPase mitofusin 2 cause Charcot-Marie-Tooth neuropathy type 2A. Nat. Genet 36, 449–451. 10.1038/ng1341 [DOI] [PubMed] [Google Scholar]

RESOURCES