ReviewMitophagy, Autophagy and Cell Death

Autophagy, apoptosis, and mitochondria: molecular integration and physiological relevance in skeletal muscle

Published Online:https://doi.org/10.1152/ajpcell.00261.2018

Abstract

Apoptosis and autophagy are processes resulting from the integration of cellular stress and death signals. Their individual importance is highlighted by the lethality of various mouse models missing apoptosis or autophagy-related genes. In addition to their independent roles, significant overlap exists with respect to the signals that stimulate these processes as well as their effector consequences. While these cellular systems exemplify the programming redundancies that underlie many fundamental biological mechanisms, their intertwined relationship means that dysfunction can promote pathology. Although both autophagic and apoptotic signaling are active in skeletal muscle during various diseases and atrophy, their specific roles here are somewhat unique. Given our growing understanding of how specific changes at the cellular level impact whole-organism physiology, there is an equally growing interest in pharmacological manipulation of apoptosis and/or autophagy for altering human physiology and health.

INTRODUCTION

Depending on their status, stress signal integration will direct cellular response mechanisms, ultimately leading to survival or death. Many forms of regulated cell death (RCD) exist, allowing cells to appropriately mediate their own destruction. Apoptosis is a RCD process characterized by highly structured and well-conserved molecular signaling events that typically serves as the default cell death pathway. Currently attributed with both prosurvival and prodeath functions, autophagy is an intracellular degradation mechanism that activates in response to various stimuli. Interestingly, on a cellular level, several stressors induce autophagy before or during apoptotic processes. While initial assessments of these observations concluded that autophagy contributed to cellular demolition and elimination, subsequent analyses indicate that autophagy typically functions to mitigate the encountered stress. Although autophagy generally appears to enhance cell survival, it is likely that an optimal level exists and that specific conditions of overactive and underactive autophagy can lead to cell death or be pathological.

Although numerous forms and condition-specific cell death mechanisms exist, including necrosis, necroptosis, ferroptosis, pyroptosis, parthanatos, entotic cell death, NETotic cell death, and immunogenic cell death (68), this review outlines the molecular signals that mediate apoptosis and autophagy, highlighting those involved with regulating both. Moreover, we focus on mitochondrial apoptotic signaling and mitophagy, due to mitochondria’s critical role in cell life and death. We also describe a general framework for the apoptosis/autophagy relationship and use several cellular mechanisms to explain how this unfolds. Finally, the roles that apoptotic and autophagic interactions play during pathophysiological conditions in skeletal muscle and other tissues are highlighted.

MITOCHONDRIAL APOPTOTIC PROCESSES

Apoptosis

Apoptosis is a physiological RCD mechanism responsible for eliminating abnormal, damaged, and/or unnecessary cells (68, 85, 138, 224). During development, specific cells undergo apoptotic cell death, thus regulating tissue/organ shape and function (224). In adult organisms, apoptosis is responsible for removing damaged and/or genetically disrupted cells as well as those affected by pathogens (138, 224). Apoptosis represents a relatively clean method of cell death that is considered almost immunologically silent (280).

The primary executioners of apoptosis are a family of enzymes known as caspases (CASPs), which cleave proteins between cysteine and aspartic acid residues (42, 68, 138, 224) (Fig. 1A). Effector CASPs (CASP3, -6, and -7) are activated by initiator CASPs (CASP8 and -9) and are responsible for cleaving >400 cellular proteins, thereby disassembling the cell (64, 169). The extracellular/extrinsic apoptotic pathways involve activation of formal death receptors embedded in the plasma membrane from the TNF receptor super family (TNFRSF) by their respective ligand (68, 82, 131) and activate CASPs using well-coordinated membrane-bound protein scaffolds.

Fig. 1.

Fig. 1.Overview of apoptosis (A) and autophagy (B) signaling pathways. A: in response to various stimuli (gray box), several signaling families [tumor necrosis factor (TNF), Jun N-terminal kinases (JNK)] activate death-associated transcription factors [tumor protein P53 (TP53)] and effector signaling mechanisms [reactive oxygen species (ROS)] (green boxes). At the mitochondria, prodeath proteins (burgundy boxes) such as BCL2-associated agonist of cell death (BAD), BH3-interacting domain death agonist (BID), BCL2-binding component 3 (BBC3/PUMA), BCL2-associated X, apoptosis regulator (BAX), and BCL2 antagonist/killer 1 (BAK1) promote mitochondria permeability, thus causing the release of cytochrome c, somatic (CYCS), diablo IAP-binding mitochondrial protein (DIABLO/SMAC), apoptosis-inducing factor mitochondrial-associated 1 (AIFM1/AIF), and endonuclease G (ENDOG). Once released, these proteins directly fragment DNA or activate caspase (CASP)9 (red boxes), ultimately promoting apoptosis. The binding of extracellular ligands at death receptors causes CASP8 (red boxes) activation through protein scaffolds [Fas-associated with death domain (FADD), TNFRSF1A associated via death domain (TRADD)]. At the endoplasmic reticulum (ER), misfolded proteins (brown clouds) and calcium mishandling cause CASP12 and calpain (CAPN) (red boxes) activation. These mechanisms are significantly redundant (dashed lines). B: numerous stimuli (gray box) affect autophagy induction by activating [AMP-activated protein kinase (AMPK), forkhead box O3 (FOXO)] and/or inhibiting [AKT serine/threonine kinase (AKT), mechanistic target of rapamycin kinase (MTOR)] stress and energy/growth factor signaling (green boxes). Beclin 1 (BECN1) and unc-51 like autophagy activating kinase (ULK) complexes (BECN1-C and ULK-C; pink boxes) mediate autophagosome formation by modulating downstream autophagy related (ATG) proteins [ATG7, microtubule-associated protein 1 light chain 3 (MAP1LC3/LC3); blue boxes], including membrane incorporation of LC3II. Autophagic substrates are identified by sequestosome 1 (SQSTM1) binding to misfolded proteins (brown clouds) with ubiquitin tags (Ub), while mitochondria are identified by unique ubiquitin binding and complex kinase-dependent activation of parkin RBR E3 ubiquitin protein ligase (PRKN) and PTEN-induced kinase 1 (PINK1) (purple boxes). BCL2-interacting protein 3 (BNIP3), BCL2-interacting protein like (BNIP3L/NIX), FUN14 domain-containing 1 (FUNDC1), optineurin (OPTN), calcium-binding and coiled-coil domain 2 (CALCOCO2/NDP52), and cardiolipin (CL) (purple boxes) also identify mitochondria to be degraded. After lysosomal fusion, autophagosome contents are degraded by proteolytic enzymes. These peptides/amino acids are released into the cytosol for recycling or used for immunomodulatory functions.


Intracellularly, stressors sensed by various compartments induce apoptosis. Mitochondria are vital mediators of many apoptotic programs whereby toxic stimulants, growth factor exhaustion, DNA damage, and reactive oxygen species (ROS) disrupt electron transport, ATP production, and mitochondrial permeability, ultimately causing release of cell death signaling proteins into the cytosol (Fig. 1A) (68, 85, 138, 224). The BCL2 apoptosis regulator (BCL2) family plays a critical role in regulating these signals through their antiapoptotic and proapoptotic functions. Upon their activation or increased expression, members such as BCL2-associated X, apoptosis regulator (BAX) and BCL2 antagonist/killer 1 (BAK1) bind antiapoptotic members such as BCL2 and BCL2 like 1 (BCL2L1/BCLXL) (37, 125, 126, 143, 151, 200), thereby permeabilizing mitochondrial membranes (4, 22, 80), reducing membrane potential, and preventing ATP generation (62, 138). This causes release of cytochrome c, somatic (CYCS) and diablo IAP-binding mitochondrial protein (DIABLO/SMAC), which contribute to CASP activation, as well as release and translocation of apoptosis inducing factor mitochondrial-associated 1 (AIFM1/AIF) and endonuclease G (ENDOG), which directly causes DNA fragmentation (51, 138, 154, 155, 224, 251). These mechanisms are highly redundant and feedforward, ensuring apoptotic execution upon reaching a certain threshold of mitochondrial damage.

Mitochondrial Permeability Transition

Few aspects of cell death have generated more controversy than mitochondrial permeability transition pore (mPTP) formation (14, 18, 59, 75, 100, 113, 138). The existence of mPTP, or a transition to increased mitochondrial permeability, is widely accepted. mPTPs occur in response to severe oxidative stress or cytosolic ion overload through assembly of large protein channels that span both mitochondrial membranes (16, 18, 138). Physiological relevance for mPTP has been recognized for several years (15). In fact, clinical trials involving its pharmacological manipulation have been conducted using cyclosporine A (CsA) to inhibit peptidylprolyl isomerase D (PPID/CYPD) immediately following or during myocardial infarction (45, 181, 210, 281). Despite demonstration in rodent models that CsA administration or PPID inhibition prevents tissue damage and increases survival during ischemia/reperfusion (IR) in cardiac and neural tissues (77, 110, 160, 223), the final clinical data suggest that this is not as significant in humans (45, 161, 281). This is potentially due to the incomplete picture regarding mPTP formation and function. While PPID is considered a component and regulator of mPTP, several other proteins previously suggested to function similarly have been subsequently identified as irrelevant or unnecessary, including components of ATP synthase (1, 59, 75, 99, 100), voltage dependent anion channels (VDACs) (11, 289), and solute carrier family 25, member 4/5/6 (SLC25A4/5/6; ANTs) (19, 89, 134), among others (68, 138).

MOLECULAR REGULATION OF AUTOPHAGY

Overview

Autophagy is a degradative process responsible for breaking down subcellular content (2, 98, 152, 215). Autophagy is uniquely flexible given that it can degrade specific targets, entire organelles, and large portions of cytoplasm. Autophagy operates by generating double-membrane organelles, filling them with cargo, and fusing them with lysosomes, where their contents are degraded and recycled (Fig. 1B) (98, 152, 215). Autophagy’s primary function is to sacrifice cellular material to provide energetic substrates during periods of starvation. As such, systemic deletion of many autophagy-related genes is lethal in mice, largely due to metabolic stress during the transition from in utero feeding (63, 135, 141, 225, 226, 246). However, autophagy is additionally involved in defense, remodeling, and removal of damaged and long-lived proteins and organelles. Given this, autophagy is generally considered cytoprotective, serving to prolong optimal cellular function (2, 98, 152, 215). Several classifications of autophagy exist; however, herein the mechanism of macroautophagy involving autophagosome-lysosome fusion will be referred to as autophagy (98, 152).

Initiation

Autophagy begins with the production of a double membrane structure known as the isolation membrane by two kinase complexes (98, 215). One complex contains unc-51 like autophagy activating kinase 1/2 (ULK1/2), autophagy related (ATG)13, and RB1-inducible coiled-coil 1 (RB1CC1/FIP200) (2, 98, 215). Under nutrient-rich conditions, the mechanistic target of rapamycin complex 1 (MTORC1) is active and exists in close association with ULK1/2 (2, 107). In this state, MTORC1 maintains ULK1/2 and ATG13 hyperphosphorylation, thereby inhibiting them (107, 119). Mechanistic target of rapamycin kinase (MTOR) inhibition causes its dissociation from ULK1/2, leading to ULK complex translocation to the site of isolation membrane production, where it activates other autophagy-related machinery (70, 107, 120). The second kinase platform is composed of beclin 1 (BECN1), phosphatidylinositol 3-kinase catalytic subunit type 3 (PIK3C3/VPS34), and phosphoinositide-3-kinase regulatory subunit 4 (PIK3R4/VPS15) as well as other components such as ATG14 and autophagy and beclin 1 regulator 1 (AMBRA1) (98, 215). This complex is a class III phosphatidylinositol 3-kinase (PI3K), and produces phosphatidylinositol 3-phosphate (PI3P) for the autophagosome membrane (257). The phosphatidylinositol (PI) for this reaction is sourced from other membranes, such as the plasma membrane (214), ER (95, 96), or mitochondria (88). ER-mitochondria contact sites, known as mitochondrial-associated membranes (MAMs), are the likely source of isolation membranes formation, as upstream autophagy-related proteins converge here during autophagy induction (71, 92). Ultimately, PI3P induces autophagosome development by recruiting adaptor proteins responsible for double-membrane production (9, 212).

Elongation and Execution

Autophagosomes are produced by ULK and BECN1 complexes with the help of two ubiquitin-like conjugation systems. The first involves a complex composed of ATG12, ATG5, and ATG16 (93, 183) that is activated by ATG7 in an E1-like manner. The second system is responsible for activating an important member of autophagosomes, microtubule-associated protein 1 light chain 3 (MAP1LC3/LC3). LC3 is a cytosolic protein whose autophagic role is initiated by ATG4-dependent cleavage, leaving a product known as LC3I (101, 255). LC3I is similarly activated by ATG7 and conjugated to phosphatidylethanolamine (PE) by the E2-like carrier ATG3 (121). The LC3I-PE conjugate, termed LC3II, is recruited by the ATG5-ATG12-ATG16 complex to the developing isolation membrane. Once the proper molecular machinery has been recruited, the ULK and BECN1 complexes guide autophagosome elongation in association with ATG5-ATG12-ATG16, making LC3II a major membrane component (121, 255). Upon their completion, autophagosomes fuse to lysosomes with the help of lysosomal associated membrane proteins (LAMPs), forming a structure known as the autolysosome (254). Finally, lysosomal hydrolases and cathepsins break down the autophagic cargo as well as the inner autophagosome membrane, including LC3II (98, 215). Degraded material is released into the cytosol, where it is used for energy metabolism, protein synthesis, and various other tasks (98, 152, 215).

Targeting

Autophagic substrates can be identified through the interaction of LC3II with the multifunctional adaptor protein sequestosome 1 (SQSTM1/P62), which is commonly found in protein aggregates (98, 215). While damaged and misfolded proteins tagged by ubiquitin can be targeted by the proteasome, SQSTM1 can also identify mono- and polyubiquitinated proteins and directly bind to them via its ubiquitin-associated domain (UBA) (109, 205). SQSTM1 subsequently binds to LC3II, thus directing specific substrates for autophagic degradation (109, 205). This function is vital not only for the clearance of accumulated proteins but for basal autophagy, and therefore, SQSTM1 is commonly analyzed as an indicator of autophagic flux (133). NBR1 autophagy cargo receptor (NBR1) functions similarly and additionally binds directly to SQSTM1, where they act together as autophagy receptors (130). Another method of aggresome degradation involves the recruitment of heat shock proteins and the E3-ligase STIP1 homology and U-box containing protein 1 (STUB1/CHIP) along with the cochaperone BCL2 associated athanogene 3 (BAG3) (32, 69). Here, misfolded and ubiquitinated targets are sequestered by heat shock complexes and shuttled to developing autophagosomes through the interaction between BAG3, SQSTM1, and LC3 (32, 69).

Control

Because overactive autophagy would be unnecessarily catabolic, its execution is precisely regulated. MTORC1 is perhaps the most important autophagy control point (2, 98, 215). Active MTOR phosphorylates specific targets, resulting in promotion of protein synthesis and prevention of autophagy; therefore, its inhibition reduces mRNA translation and induces autophagy (215). Insulin, glucagon, and other growth factor signaling affect autophagy through an AKT serine/threonine kinase (AKT)-MTOR axis. Further, autophagy is highly sensitive to alterations in energy/substrate availability. For example, when amino acids are available, MTORC1 activity is promoted via the interaction between regulatory associate protein of MTOR complex 1 (RPTOR/RAPTOR) and the RAG family of small GTPases (124, 230). Therefore, reduced cellular amino acid concentrations caused by starvation remove the Ras homolog, MTORC1 binding (RHEB)-mediated sensitization of MTOR to nutrient availability, releasing the brake on autophagy induction (229). Similarly, nutrient unavailability increases the AMP/ATP ratio, resulting in AMP-activated protein kinase (AMPK) activation. AMPK inhibits MTORC1 by phosphorylating TSC complex subunit 2 (TSC2) and RPTOR (86, 111). Furthermore, AMPK can activate autophagy by directly phosphorylating ULK1/2 (128, 149).

MITOPHAGY AS TARGETED AUTOPHAGY

Overview

Mitophagy is a process often regulated independently of the nutrient/energy/stress signals that govern basal autophagy (7). It operates primarily as a quality control mechanism, targeting dysfunctional mitochondria that may otherwise contribute to the activation of death signaling (7). Additionally, mitophagy is responsible for eliminating healthy mitochondria during the differentiation of several cell types (10, 43, 144, 231, 242). While the stimuli for mitophagy and “nonspecific” autophagy may differ, mitochondria are degraded by autophagosome sequestration and subsequent lysosomal fusion (Fig. 1B). In this sense, mitochondria are simply treated as very large autophagic substrates, thereby requiring activation of similar autophagosome molecular machinery (i.e., BECN1, ULK, LC3, etc.) (7). However, because of the size and complexity of mitochondrial networks, specific mitophagy events related to mitochondrial identification and sequestration exist.

PINK1 and PRKN

Mitophagy involves unique and additional substrate identification mechanisms, notably PTEN-induced kinase 1 (PINK1) and parkin RBR E3 ubiquitin protein ligase (PRKN), two genes whose mutations were initially associated with recessive forms of parkinsonism (132, 263). Normally, PINK1 is constitutively transported into mitochondria, cleaved by the protease presenilin associated rhomboid like (PARL) , and degraded by mitochondrial proteases (49, 117). When mitochondria become dysfunctional, PINK1 degradation is impaired, and it stabilizes on the outer mitochondrial membrane (117, 192, 193). In response, PRKN translocates to sites of mitochondrial damage denoted by PINK1 presence (73, 117, 192, 193). It is thought that mitochondrial proteins phosphorylated by PINK1 serve as PRKN docking sites or that direct phosphorylation of PRKN stimulates its translocation (7, 129). The phosphorylation of ubiquitin by PINK1 and formation of specific polyubiquitin chains has also been shown to regulate PRKN recruitment (240).

PRKN ubiquitinates outer mitochondrial membrane proteins following its translocation, causing mitochondrial sections to be isolated (36, 72). These fragments are identified by ubiquitin-SQSTM1-LC3 autophagosome targeting and degraded following lysosomal fusion (53, 73, 191). However, reports have indicated that SQSTM1 is required for mitochondrial clustering/fragmentation, but not in their degradation specifically (147, 191, 197). Several proteins identified as PRKN substrates appear to be important for mitochondrial fragmentation, including mitofusins (MFNs), translocases of outer mitochondrial membrane (TOMs), and VDACs (36, 250, 270, 282). PRKN-mediated MFN ubiquitination is thought to prevent defective mitochondria from fusing back into the mitochondrial network, although cells lacking MFNs can undergo mitophagy (36, 253). Importantly, excessive fusion caused by overexpression of OPA1 mitochondrial dynamin like GTPase (OPA1) or dominant-negative dynamin 1 like (DNM1L/DRP1) inhibits mitophagy (261). Thus, functioning mitochondrial fission/fusion machinery should be considered a prerequisite for mitophagy. VDACs are also required for efficient PRKN recruitment, and their ubiquitination is necessary for mitophagy (73, 250). PRKN also ubiquitinates TOMs during membrane depolarization, stimulating their degradation and permitting PINK1 accumulation (146, 282).

BNIP3 and BNIP3L

These BCL2 family members possess autophagy-promoting capabilities independent of their proapoptotic functions associated with being BH3-only proteins (91, 173, 218, 259, 285). BCL2-interacting protein 3 (BNIP3) and BCL2 interacting protein like (BNIP3L/NIX) induce autophagy by competitively binding to and displacing BCL2 and BCL2L1 from BECN1 and perhaps by depolarizing mitochondria (13). Furthermore, both interact directly with LC3 via LC3-interacting region (LIR) domains and are important for autophagosome-mitochondria targeting, potentially inducing mitophagy independent of PINK1 and PRKN (94, 195, 218, 238). In fact, BNIP3 overexpression can compensate for PINK1 deficiency and reverses skeletal muscle degeneration in this context (286). Alternatively, although PINK1- or PRKN-mediated exposure of BNIP3 and BNIP3L LIR domains is enticing, such interactions have not been characterized. However, BNIP3 affects PINK1 function by suppressing its proteolytic cleavage, thus enabling PINK1-dependent mitophagy during hypoxia (286). In separate studies performed in cardiomyocytes, BNIP3 and BNIP3L triggered DNM1L translocation to mitochondria, resulting in mitochondrial fission followed by PRKN-dependent mitophagy (53, 150).

Other Mitophagy Receptors

Several molecules aid autophagic identification of mitochondria. For example, BCL2 like 13 (BCL2L13/BCL-RAMBO) was demonstrated to be required for mitochondrial fragmentation and mitophagic degradation in HEK293 cells (189). This protein was capable of causing mitochondrial fragmentation independent of DNM1L through its BH domains, binding to LC3 using its LIR domains, and incorporating mitochondrial fragments into lysosomes and depleting mitochondrial proteins in the absence of PRKN. However, because of their relative complexity, previous research in mammalian cells has characterized a growing list of additional molecules that regulate mitochondrial targeting and mitophagy during various conditions, including SQSTM1, NBR1, FUN14 domain containing 1 (FUNDC1), AMBRA1, cardiolipin, Tax1 binding protein 1 (TAX1BP1), calcium-binding and coiled-coil domain 2 (CALCOCO2/NDP52), optineurin (OPTN), TANK-binding kinase 1 (TBK1), and FKBP prolyl isomerase 8 (FKBP8) (20, 21, 41, 63, 102, 130, 147, 164, 189, 195, 217, 218, 248).

Despite the array of mitophagy-related adaptors, OPTN and CALCOCO2 have been identified as the de facto receptor proteins required for mitophagy (102, 147, 217). These two receptors function partly redundantly to link phosphorylated polyubiquitin to LC3 and subsequent PRKN-dependent enhancement of mitochondrial membrane protein ubiquitination (147). Here, PINK1-dependent phosphorylation of ubiquitin is vital to activating ubiquitin, OPTN, and CALCOCO2, irrespective of PRKN presence. TBK1-dependent phosphorylation of OPTN, CALCOCO2, TAXBP1, and SQSTM1 in turn creates an amplification loop linking ubiquitinated receptors to autophagosomes (217). Importantly, PRKN-independent execution of mitophagy has also been described (249). For example, AMBRA1 promotes mitochondrial sequestration and mitophagy in PRKN-expressing cells; however, in the absence of PRKN and SQSTM1, AMBRA1 relocated to depolarized mitochondria and recruited LC3; a function dependent on its LIR (249). Similar observations of PRKN- (20, 156, 189, 245) and PINK1-independent (139) mitophagy have also been made by others. The existence of multiple and redundant mitochondrial targeting mechanisms underscores the importance of accurate substrate identification in mitophagy.

Mitophagy Control

As mitophagy requires autophagosome sequestration, several feedback mechanisms link to autophagy initiation. BECN1 is involved with mitochondrial translocation of PRKN, where it colocalizes with PINK1 and PRKN at MAMs (39, 74). Here, PINK1 aids in recruiting BECN1 to MAMs independently of PRKN, and this is required for proper MAM-associated autophagosome biogenesis (74). AMBRA1 traffics to depolarized mitochondria and promotes isolation membrane production and mitochondrial clearance (249, 264). Full-length PINK1, which would accumulate only during mitochondrial depolarization, also interacts with BECN1 to promote autophagy (182). BNIP3 and BNIP3L expression is similarly stress-induced by hypoxia through hypoxia inducible factor 1 subunit-α (HIF1A) (13) and by starvation through forkhead box O3 (FOXO3) (173). Finally, CALCOCO2 and OPTN phosphorylation activates upstream autophagy-regulating proteins (147). Therefore, it appears that mitochondria prepped for mitophagy are degraded alongside other autophagy substrates, and this occurs largely when markers of dysfunction (PRKN, ubiquitin, SQSTM1, PINK1, etc.) are present.

DEFINING THE RELATIONSHIP BETWEEN APOPTOSIS AND AUTOPHAGY

Delineating the mechanisms and functions of stress response processes is complex, as they depend on interacting stress- (type and duration) and cell-specific (type and status) variables. Although the mechanisms underlying the relationship between apoptotic signaling and autophagy are complex, their consequences can be summarized as follows. 1) In the absence of stress, autophagy operates at a basal level, thereby providing important cellular functions, while apoptotic signaling is kept to a minimum; 2) relatively low-level stress induces autophagy as well as activates some antiapoptotic mechanisms to resist or remove stress sources; 3) if deemed unsurvivable, unabated stress activates proapoptotic mechanisms, thereby disabling autophagy to ensure cell elimination in a physiologically friendly manner (68, 79, 174, 265) (Fig. 2A). This is typically due to the attenuation of cellular stress by autophagy (Fig. 2B) and the subversion of autophagy by apoptotic mechanisms (Fig. 2C). As indicated by the numerous studies cited in this review, this dose-response relationship holds true for most cellular stressors in addition to nutrient starvation, including foreign chemicals, nonbiological stresses (heat, pressure), extracellular physiological stress signals (cytokines, DAMPs), changes to biologically relevant conditions (pH, redox), metabolic alterations (substrates, metabolic intermediates), external growth status factors/hormones, viral and bacterial infection, internal organelle dysfunction, and DNA damage; although, the degree of apoptosis to autophagy varies widely across these stimuli.

Fig. 2.

Fig. 2.General relationship between autophagy and apoptosis. A: most stresses progressively induce autophagy and apoptosis. Furthermore, cells with larger autophagy responses display relative resistance to apoptosis, suggesting that higher sensitivity to autophagy induction causes resistance to that stress. Typically, autophagy induction occurs at relatively low stress levels, followed by activation of apoptosis as intensity and/or duration increases. B: autophagy is generally cytoprotective by removing stress sources. In this example, mitochondrial dysfunction eventually leads to permeabilization, reactive oxygen species (ROS) generation, release of prodeath stimuli [i.e., cytochrome c, somatic (CYCS)], caspase (CASP) activation, and apoptosis. However, degradation of impaired mitochondria removes this potential stress source and allows for the generation of new mitochondria. C: prolonged activation of apoptotic effectors eventually subverts stress-induced autophagy by cleaving numerous autophagy-related proteins [beclin 1 (BECN1), parkin RBR E3 ubiquitin protein ligase (PRKN), autophagy and beclin 1 regulator 1 (AMBRA1), autophagy related (ATG)3, ATG4, and ATG5]. The BECN1, ATG5, and ATG4 cleavage products additionally acquire prodeath functions, reinforcing cell death. D and E: experiment demonstrating cytoprotective and dose-response nature of autophagy and apoptosis during various starvation conditions. D: we generated 3 ATG7-deficient clonal mouse myoblast (C2C12) cell lines using shRNA (shAtg7) as well as a scramble control line (SCR). E: these cells were subjected to various starvation modes. shAtg7 cells displayed significantly elevated CASP3 activity that was highly time and growth factor dependent. *Significant difference between the denoted group and C2C12 normal culture growth media [GM; DMEM (DM) with 10% FBS] calculated using a t-test; P < 0.05. GM/HB or DM/HB, 1:1 mixture; HB, Hanks’ Balanced Saline Solution; 1%/5%, percentage of FBS. HB contains similar glucose as DM but contains no amino acids.


We approach describing how the stimuli, mechanisms, and consequences between apoptotic signaling and autophagy interact assuming such a model. Because separating the mechanisms that allow communication between apoptosis and autophagy from their stimuli is difficult, for simplicity we have separated these explanations by stimuli/mechanisms as appropriate.

MECHANISMS UNDERLYING THE APOPTOSIS/AUTOPHAGY RELATIONSHIP

Overview

The sequential induction of autophagy and apoptosis likely allows cells to mediate damage using autophagy before reaching a level obliging death (7). In fact, cells that mount the most robust autophagic response display the greatest apoptotic resistance (278). This cytoprotection is routinely demonstrated, where induction of autophagy prevents apoptosis or inhibition of autophagy promotes apoptosis (174). Given that this response is well established and beyond the scope of this review, we will focus on describing a number of key cellular processes shared by autophagy and apoptosis, as well as how these diverse factors influence the autophagy/apoptosis response.

Starvation and Growth Factor Signaling

Relative starvation robustly induces autophagy in cultured cells and whole organisms (184, 186, 228). However, prolonged starvation leads to both death of cells [through programmed signaling pathways (241)] and organisms. Studies showing that autophagy inhibition induces apoptosis during starvation were performed early during its examination in mammalian cells (25). Moreover, starvation-sensitive decisions regarding autophagy and apoptosis are influenced by growth factor receptor signaling in a dose-dependent manner. For example, in a small experiment presented here, we clearly observed elevated CASP3 activity in autophagy-deficient cells during starvation that was growth factor and time dependent (Fig. 2, D and E). In apoptosis-resistant Bax−/−/Bak1−/− cells, autophagy helps prolong viability during extended starvation, and this is sensitive to extracellular growth factors (167). Mechanistically, Jun N-terminal kinases (JNK)-dependent phosphorylation of BCL2 decreases its inhibition of BECN1, thereby permitting autophagy induction during starvation (272). JNK signaling also contributes to mitophagy by inducing FOXO3-dependent transcription of Bnip3 (35). Similarly, extracellular signal-regulated kinases (ERK) and MAPK kinases (MEK) inhibition partially and completely, respectively, prevents autophagy during starvation and in response to rapamycin (269). However, it is also well established that ERK and JNK signaling events activate apoptosis (29, 52). ERK-dependent apoptosis occurs in response to numerous stimuli, particularly DNA-damaging chemicals and extracellular ligands, and involves CASPs, BCL2, and tumor protein P53 (TP53) (29). Notably, these responses are typically observed during prolonged (≥24 h) or constitutive stimulation (29, 52).

Mitochondria

Mitochondria are central regulators of apoptosis and autophagy. Briefly, mitochondria contribute to the stress-response relationship because dysfunctional mitochondria (i.e., with decreased membrane potential) are preferentially targeted for mitophagy, thereby removing them as potential sources of cellular damage (Fig. 3A) (65, 127, 192, 193, 261, 271). Left unabated, mitochondria become increasingly permeabilized, pores form across their membranes, and feed-forward apoptotic processes reinforce the release of death-inducing proteins and collapse of ATP production (138). Mitophagy has repeatedly been shown to limit mitochondrial ROS production (142, 235), but additional mechanisms have been observed, including attenuation of CYCS release and CASP3 activity (279). Reducing PRKN protein levels sensitized neural cells to apoptotic cell death (170), while overexpression protected cardiomyocytes from apoptotic death during hypoxia (140). BNIP3-mediated mitophagy also limits mitochondrial amplification of apoptosis by reducing CYCS release (290). In addition to sequestering dysfunctional mitochondria, other actions associated with mitophagy prevent apoptosis. Upon mitochondrial depolarization, PINK1 stabilizes the antiapoptotic abilities of BCL2L1 by phosphorylating and preventing its cleavage (6). Although cytosolic TP53 can bind to and inhibit mitochondrial PRKN translocation, PRKN prevents TP53-induced CASP3 activation by acting as a transcriptional repressor of TP53 (46). Cardiolipin externalization also contributes to mitochondrial targeting by binding LC3 (41) in addition to promoting CYCS release when oxidized (202).

Fig. 3.

Fig. 3.Examples of shared stress response signaling mechanisms that allow interaction and tuning of apoptosis and autophagy. A: mitochondrial damage [i.e., via reactive oxygen species (ROS), mtDNA mutations, or mitochondrial permeability transition pore (mPTP)] causes membrane depolarization and recruits mitophagy machinery [PTEN-induced kinase 1 (PINK1) and parkin RBR E3 ubiquitin protein ligase (PRKN)]. Unchecked or widespread mitochondrial damage leads to permeabilization, release of death signaling factors [cytochrome c, somatic (CYCS), diablo IAP-binding mitochondrial protein (DIABLO/SMAC), apoptosis inducing factor mitochondrial-associated 1 (AIFM1/AIF)], caspase (CASP) activation, and apoptosis. B: anti-death BCL2 apoptosis regulator (BCL2) family members [BCL2, BCL2 like 1 (BCL2L1/BCLXL)] inhibit autophagy by physically binding to beclin 1 (BECN1). BH3-only members [BCL2 binding component 3 (BBC3/PUMA), BH3-interacting domain death agonist (BID), BCL2-associated agonist of cell death (BAD)], which increase or are activated in response to stress, sequester BCL2, thereby relieving its inhibition of BECN1. Eventually, BCL2 becomes saturated with BH3-only proteins, leading to BCL2 antagonist/killer 1 (BAK1) and BCL2-associated X, apoptosis regulator (BAX) activation and apoptosis. C: ROS (O2, H2O2) can be scavenged by various antioxidants [superoxide dismutase (SOD), glutathioine (GSH), catalase (CAT)] and are important second messengers and regulate numerous cellular processes [extracellular signal-regulated kinases (ERK), mitogen-activated protein kinases (MAPK), sirtuins (SIRT), AMP-activated protein kinase (AMPK), nuclear factor-κB (NFKB), PPARG coactivator-1α (PPRAGC1A/PGC1α), nuclear factor, erythroid 2 like 2 (NFE2L2/NRF2)]. With respect to autophagy, elevated H2O2 is required to activate autophagy related 4 (ATG4), which in turn participates in LC3 maturation. Unrestrained ROS production activates death-inducing signaling mechanisms [tumor protein P53 (TP53), Jun N-terminal kinases (JNK), mitogen-activated protein kinase kinase kinase 5 (MAP3K5/ASK1)] and damages lipids, DNA, and proteins, ultimately reinforcing apoptosis.


BCL2 Proteins

BCL2 family proteins constitute a straightforward explanation for the apoptosis-autophagy relationship. As BECN1 contains a BH3 domain, activity of the BECN1 complex is reduced by physical interactions between antiapoptotic BCL2 and BCL2L1 with BECN1; accordingly, BH3-only proteins bind to BCL2 and BCL2L1, freeing BECN1 and thus promoting autophagy (Fig. 3B) (172, 207). BNIP3 and BNIP3L similarly interrupt BECN1-BCL2 binding in addition to functioning as mitophagy receptors (13, 94, 195). Notably, BH3-only proteins exist under strong transcriptional and posttranslational control by death-inducing and stress-sensing mechanisms (68). Similarly, BNIP3 and BNIP3L expression is hypoxia inducible (13). When considered as a model, 1) in the absence of stress, BCL2 and BCL2L1 bind to BECN1 and restrain autophagy, 2) stress activates BH3-only proteins which replace BECN1, thereby allowing BECN1 complex activation, and 3) prolonged stress increases BH3-only protein levels above the absorbing threshold of BCL2 and BCL2L1, causing BAX and BAK1 activation, widespread mitochondrial permeabilization, CASP activation, and apoptosis. The mitochondrial location of BCL2 proteins suggests that BH3-only proteins and BAX and BAK1 may contribute to mitochondrial depolarization and mitophagy at low activation levels but promote apoptosis during prolonged stress.

Reactive Oxygen Species

ROS also illustrate this dose-response relationship (Fig. 3C). Despite their connection to apoptosis, cancer development, and cellular aging, ROS are vital second messengers that execute various functions (237). This includes augmenting growth factor signaling, activating metabolic enzymes, enhancing the inflammatory response, increasing transcription of protective and antioxidant genes, and mediating long-term metabolic adaptations (237). Although ROS induce autophagy by causing cellular damage that autophagy attempts to mitigate, ROS also directly regulate autophagy (152, 174, 215). In fact, antioxidant administration depresses starvation- (236) and ROS-induced (30, 163) autophagy. Specifically, oxidation of ATG4 by H2O2 is required for autophagosome production (236), ROS initiate PRKN-dependent mitophagy (271), and ROS-induced inhibition of MTOR is AMPK dependent (3). In this latter paper, ROS-induced autophagy required the DNA damage sensor ATM serine/threonine kinase (ATM), and rescuing autophagy in ATM-deficient mice by administering rapamycin reversed ROS-induced lymphomagenesis (3). Typically, autophagy protects from ROS-induced apoptosis (174, 215, 235). Despite this, in specific cell types suppression of autophagy-related genes actually ameliorates ROS-induced apoptosis (38), demonstrating the complexity of this relationship. However, eventually, cellular ROS-mitigating mechanisms become saturated, preventing their conversion to H2O2 and H2O and causing highly reactive superoxide and hydroxyl radicals to accumulate (237). In addition to actively promoting programmed death mechanisms (i.e., TP53 and JNK), these species nonspecifically damage DNA, proteins, and lipids, thereby promoting apoptosis (243).

Apoptotic Enzymes

As previously alluded to, apoptotic signaling mechanisms are graded and do not automatically cause cell death. Particularly, CASPs participate in the differentiation of various epithelial tissues, erythrocytes, and skeletal muscle (62). Although evidence of CASP-dependent autophagy at relatively low activation levels is uncommon, one study showed that CASP9 was required for cytoprotective autophagy induced by an anti-inflammatory chemical in MCF7 cells (115). However, effector CASPs eventually advance apoptotic execution, partly by subverting autophagy (Fig. 2C). CASP3 cleaves BECN1 during apoptosis induced by BAX, thereby inhibiting autophagy (168). In fact, the COOH-terminal fragment of BECN1 translocates to mitochondria, where it causes membrane permeabilization (168, 275). AMBRA1 is also cleaved by CASPs and calpains (CAPNs) during staurosporine induced apoptosis, subsequently contributing to inhibition of autophagy (203). Similarly, the BECN1 complex member ATG3 undergoes CASP cleavage during apoptotic stress (201). Substrates for apoptosis-induced inactivation of autophagy apart from the BECN1 complex have also been identified. Of note is that CAPN-mediated ATG5 cleavage sensitizes tumor cells to apoptotic stress and produces a cleavage product that undergoes mitochondrial translocation, thereby contributing to CASP activation (284). Similarly, the product of CASP cleavage of ATG4 has apoptosis-promoting effects (17). Finally, CASPs cleave and inactivate PRKN, preventing mitophagy and promoting cellular damage (122). The large number of ATGs targeted by apoptotic enzymes strongly supports the view that autophagy is an evolutionarily conserved cell survival process.

DNA Damage

Apoptosis resulting from DNA damage typically manifests in the mechanisms just described (i.e., BH3-only protein transcription, ROS generation, and CASP activation). However, the apoptosis regulator TP53 has independent autophagy-regulating roles, where it is primarily inhibitory (256). Cytosolic localization of TP53 protein allows its direct interaction with and inhibition of RB1CC1 (187) and PRKN (106), thereby reducing BECN1 complex activity and preventing mitochondrial autophagic clearance. However, during situations of induced autophagy, TP53 is responsible for activating transcription of tuberous sclerosis complex (TSC) and AMPK components, two platforms that promote autophagy (61). The TP53 transcriptional target DNA damage regulated autophagy modulator 1 (DRAM1) is also responsible for executing TP53-dependent autophagy during DNA damage and inhibition of mitochondrial respiration (44, 287). While the autophagy versus apoptotic interactions that determine these responses are relatively unknown, it is likely that the type and intensity of stress stimuli create an environment favoring one or the other following DNA damage.

Autophagy-Dependent Cell Death

Unrestrained autophagy and mitophagy would be unnecessarily catabolic and theoretically lead to mitochondrial depletion. While this is not commonly observed in mammalian cells, 1) some autophagy machinery is involved in cell death execution, and 2) some cancer cells undergo autophagy-dependent cell death (174). Although CASP8 is typically activated by the death-induced signaling complex (DISC), a similar platform forms on autophagosomes and is required for complete enzyme activation (283). ATG12 can also promote apoptosis by binding to and inhibiting BCL2, a function required for full CYCS release during staurosporine-induced apoptosis (221). Furthermore, autophagy inhibition prevents cell death induced by falcarindiol in breast cancer cells (166), by MG-2477 in neuroblastoma cells (87), and by sunitinib in prostate cancer cells (268). Similarly, adiponectin attenuated mitophagy and apoptosis induced by H2O2 in C2C12 cells (216). Interestingly, mitophagy inhibition decreased cell loss during starvation by causing mitochondrial fusion, thereby maintaining ATP production (76). However, when fission and, therefore, mitophagy were enhanced, starvation increased death (76).

PHYSIOLOGICAL IMPLICATIONS ACROSS THE APOPTOSIS/AUTOPHAGY AXIS

Overview

Importantly, the consequences of autophagy’s impact on cellular stress resistance and function are relevant to numerous pathological conditions. Due to overlapping regulatory mechanisms, defective autophagy and mitophagy contribute to unnecessary apoptosis and tissue loss, and the development of cellular proapoptotic environments that can decrease the limit of survivable stress (98, 152, 215).

Autophagy/Mitophagy and Apoptosis in Various Tissues

Numerous tissues show examples of altered mitophagy impacting their pathophysiology. For example, insufficient mitophagy is directly related to neuronal cell loss and/or dysfunction during Parkinson’s (132, 263), Alzheimer’s (12, 220), and aging (12). Pancreatic β-cells display reduced mitophagy during type 1 and type 2 diabetic conditions (104). Similarly, mitophagy in kidney proximal tubule cells is responsible for maintaining mitochondrial function during metabolic acidosis (190), whereas impaired mitophagy contributes to cell damage, such as that which occurs during diabetic nephropathy (103). In the heart, mitophagy and PRKN levels increase in the infarct border following a myocardial infarction (140), while Prkn−/− mice display larger infarct areas, ultimately demonstrating that mitophagy prevents cell loss (140). Further, mitophagy is impaired in hearts of aged Prkn−/− mice (106). In contrast, elevating mitophagy by decreasing TP53 expression is associated with increased cardiac resistance to ischemic stress (105, 106), while mice overexpressing PRKN are resistant to the age-related decline in cardiac function, and this is associated with elevated mitochondrial activity, decreased ROS production, and reduced inflammation (106). Therefore, mediation of mitochondrial-related stresses by mitophagy is necessary for preserving cell number and function.

Decreased autophagy can also contribute to tumor development (152, 273). Several common genetic alterations, such as TP53 mutations, BCL2 upregulation, and BECN1 inactivation, cause cell death avoidance and alter cell cycle while also impairing autophagy (152, 158, 177). With respect to tumor initiation, 1) autophagy impairment increases cellular stress, leading to genomic instability and oncogene activation, mediated through SQSTM1-induced nuclear factor, erythroid 2 like 2 (NFE2L2/NRF2) (58, 83, 176), and 2) without autophagy, tumor-associated stress causes immunologically noisy cell death and alters oncogenic antigen processing, leading to cancer progression (50, 152, 177, 273). Conversely, it is well established that enhanced autophagy promotes the growth of established cancers and contributes to therapy resistance (273).

Autophagy and Apoptosis in Skeletal Muscle Development

Skeletal muscle development involves dramatic morphological transformation as single-nucleated myoblasts fuse into complex and multinucleated contractile muscle fibers. Unsurprisingly, skeletal muscle differentiation is characterized by significant stress-related processes, including increased ROS production (136), MAPK signaling (277), DNA damage response (DDR) (145), CASP activation (24), mitochondrial fission (23), and autophagic (180) and mitophagic (10, 242) flux. However, despite their links to apoptosis, skeletal muscle differentiation is actually impaired by individual inhibition of many of these processes. In this context, we repeatedly observe that various modes of autophagy inhibition, including 3-MA administration (180), ATG7 knockdown (10, 180), and Bnip3 knockout (10), potentiate apoptotic signaling resulting in skeletal muscle differentiation impairment. Importantly, we found augmented mitochondrial apoptotic signaling (i.e., CYCS and AIFM1 release, increased mPTP formation, CASP9 activation) in ATG7-deficient cells (10). This suggests that autophagy induction facilitates differentiation by attenuating cellular and mitochondrial stress sources that may otherwise lead to apoptosis. In fact, we additionally demonstrated that inhibiting the augmented CASP3 or CASP9 in ATG7-deficient myoblasts recovered the differentiation ability of these autophagy-impaired cells (10). Importantly, the mechanisms of skeletal muscle formation (i.e., myoblast fusion) are similar to those regulating mature skeletal muscle adaptation, growth, and regeneration; therefore, these interactions have implications regarding the treatment of various muscle myopathies, dystrophies, and atrophies.

Autophagy in Mature Skeletal Muscle

In mature tissue, autophagy has been suggested to both contribute to and protect from specific pathological conditions (232). Primarily, autophagy degrades energetic substrates in skeletal muscle during nutrient deprivation (184, 186, 232, 288). This function is likely vital for converting and mobilizing skeletal muscle’s large protein and glycogen stores into fuel for use by other tissues. In fact, mice lacking skeletal muscle AMPK display hypoglycemia during fasting, a finding attributed to their inability to supply the liver with alanine for gluconeogenesis resulting from depressed skeletal muscle autophagy (28). However, this catabolic response must be properly regulated, and therefore, autophagy may unnecessarily contribute to atrophy during specific circumstances (232). Elevated skeletal muscle autophagic activity has been observed in response to denervation (66, 67, 84, 198, 288), fasting (184, 196, 288), exercise (97, 162), oxidative stress (54, 208), chemotherapy (26), endotoxin (114), inflammation (234), glucocorticoid administration (234, 260), and disuse (108, 252), while decreased autophagy activity has been observed in skeletal muscle during aging (116), type 2 diabetes (185), and critically ill patients (266). Although fewer examinations of specific mitophagy have been performed, indirect measurements have suggested altered mitophagy during fasting, denervation, and disuse (67, 123, 198). Additionally, PRKN-deficient mice display delayed atrophy and decreased ubiquitin proteasome system (UPS) activation during denervation (67).

Apoptosis in Mature Skeletal Muscle

Apoptotic signaling in skeletal muscle is typically associated with atrophy and dysfunction (213). Death-associated signaling mechanisms are unique in skeletal muscle, as they do not generally cause complete cell death due to its multinucleated morphology (213). Instead, CASPs cleave complex myofibrillar proteins into proteasome-compatible sizes, thereby prepping these targets for further degradation (56). The connection between apoptosis and muscle atrophy is most strikingly illustrated in Bax−/− (244), Bax−/−/Bak1−/− (198), and Casp3−/− (211) mice, which display resistance to denervation-induced atrophy. Apoptotic signaling may also damage or eliminate individual skeletal muscle nuclei and theoretically influence the cytoplasmic volume/area that could be supported and thus contribute to fiber atrophy (5). However, the myonuclear domain concept has long been questioned (27, 238) given the additional roles of satellite cells and flexibility in this ratio (188, 239). Regardless, apoptotic markers are observed in skeletal muscles of dystrophic mice (112, 258) and humans (233) as well as in lysosomal (159) and mitochondrial (8) myopathies, suggesting that apoptotic mechanisms generally contribute to pathology or indicate the existence of underlying pathological alterations.

Autophagy and Apoptosis During Skeletal Muscle Atrophy, Aging, and Pathology

Although autophagy increases during atrophy, whether this is pathological is unclear. As a catabolic process, autophagy could independently or cooperatively contribute to protein degradation and muscle loss during disuse, fasting, or stress. In our hands, skeletal muscle-specific autophagy deficiency (Atg7−/−) in the absence of additional stress increased centralized nuclei (a pathological sign of regeneration) and impaired contractility while elevating CAPN and proteasome activity in an age-dependent and muscle-specific manner (206). Interestingly, we also found elevated cytosolic CYCS and AIFM1 nuclear translocation in oxidative muscle of Atg7−/− mice, indicating generalized dysfunction and mitochondrial stress. Others have similarly shown that inactivating autophagy causes muscle dysfunction and elevates apoptotic DNA fragmentation, thereby implying that autophagy executes specific functions that protect skeletal muscle (81, 175). Furthermore, autophagy deficiency enhances atrophy and dysfunction during atrophy-inducing conditions (175), and autophagy induction during denervation does not affect atrophy (209), suggesting that autophagy’s catabolic contribution is likely not pathological. Another mouse model (miR-378KO) demonstrates impaired autophagy, mitochondrial abnormalities, muscle atrophy, and decreased running performance alongside mitochondrial CYCS release and CASP activation (157). Notably, miR-378 is dramatically induced during fasting and activates autophagy while inhibiting apoptosis by targeting CASP9 (157). The observation that chemical CASP inhibition induced autophagy and improved running performance of miR-378KO mice (157) further supports the protective role of autophagy and mitophagy against mitochondrial-associated dysfunction and apoptotic signaling.

The importance of autophagy in skeletal muscle is further highlighted by its depression during age-related atrophy, known as sarcopenia (116). Here, skeletal muscles display SQSTM1 accumulation (227, 274), depressed denervation-induced autophagy (199), downregulated autophagy gene expression (55, 118, 276), and elevated CASP activity and apoptotic nuclei (57). In this context, Prkn−/− mice display significantly impaired ADP-stimulated mitochondrial respiration and increased mPTP formation (78), while PRKN overexpression prevented aging-associated functional declines and atrophy (148); changes that were associated with maintenance of mitochondrial enzyme activity and attenuated apoptotic nuclei (148). In fact, numerous interventions, including small molecules (60, 222), caloric restriction (276) and exercise (60, 90, 267, 274, 276) attenuate skeletal muscle dysfunction and atrophy, likely due to increased autophagy and decreased apoptotic signaling.

Autophagy and Apoptosis During Skeletal Muscle Dystrophies

Several studies have shown impaired autophagic flux and elevated apoptotic signaling during several models of muscular dystrophy (31, 48, 81). Remarkably, when autophagic flux was promoted by feeding mdx mice a low-protein diet, their functional and structural abnormalities improved and the number of apoptotic nuclei decreased (48). Likewise, inducing autophagy by administering a low-protein diet, rapamycin, or spermidine decreased apoptotic markers and improved functional parameters of Col6−/− dystrophic mice (40, 81). In fact, the increased mitochondrial permeability and apoptotic nuclei seen in these mice (112) are reversed by these three autophagy inducers (40, 81), indicating an association between apoptotic signaling and pathology. Furthermore, overexpressing BECN1 independently reduced apoptotic nuclei in dystrophic mice, suggesting that autophagy induction specifically can improve pathology (81). Translating these results to humans, a low-protein diet was shown to induce autophagy and stabilize functional measures of disease progression in muscular dystrophy patients with Col6 mutations, as well as being associated with decreased apoptotic nuclei and improved mitochondrial function in skeletal muscle (33). However, emphasizing autophagy’s dual nature, in the studies just mentioned, both a long-term, low-protein diet (48, 81) and spermidine administration (40) increased apoptotic nuclei in muscle of wild-type mice, while inhibition of autophagy improved clinical symptoms of muscular dystrophy in a mouse model of MDC1A (laminin a2 chain deficiency) (31).

PERSPECTIVE ON THE PHYSIOLOGICAL RELEVANCE OF AUTOPHAGY AND APOPTOSIS

We like to characterize autophagic degradation of cellular material in a “worst is first” manner that ultimately serves to protect cells from stress. Whether these stressors stem from hazardous protein aggregates, dysfunctional organelles, ROS-damaged molecules, or invading pathogens, autophagy typically functions as one of the cell’s first lines of defense. However, in response to prolonged/high intensity stress, cells eventually capitulate to self-destruction; in this sense, apoptotic cell death would follow autophagy. Conceptually then, forced autophagy induction in the absence of additional stress preferentially degrades the most appropriate targets present at that time (i.e., relatively depolarized mitochondria, any SQSTM1-bound aggregates) despite these species not being harmful enough to autonomously induce autophagy.

This feature of autophagy likely partly explains the long-known health benefits of proper diet and regular exercise, two inducers of autophagy. In fact, genetic autophagy impairment abolishes the longevity effects of relative caloric restriction in model research organisms and animals (47, 171). Furthermore, the investigations of caloric restriction in nonhuman primates concluded that “caloric restriction without malnutrition…improves health and survival of rhesus monkeys” (178), supporting the translation of these observations to humans. Because of these findings, there is growing interest in pharmacological and lifestyle interventions that mimic caloric restriction and/or simply induce autophagy (34, 137, 153, 171, 204).

Importantly, as progress is made toward conducting a human trial investigating the effects of rapamycin on human aging (262), this means that rational design of autophagy-manipulating drugs requires deep understanding of the interplay between autophagy and apoptosis in specific cell types and in specific contexts (137). Notably, despite our expanding understanding of mitophagy, the myriad of relevant molecular mediators complicates its pharmacological targeting. For example, while ATG32 independently executes mitophagy in yeast, it is unsurprising that the complexity of mammalian cells and their relationship with mitochondria has evolutionarily optimized mitophagy by supplementing it with regulators in addition to PINK1 and PRKN. As research continues, we will likely find that defined mechanisms are relevant to individual stimuli, in the same sense that specific conditions induce ATG5/ATG7-independent autophagy (194) and mitophagy-independent lysosomal degradation of mitochondrial segments (179, 219, 247). Furthermore, as nonregulated health supplements purported to slow aging appear on the market and the trend of intermittent fasting becomes more common, determining these complexities is relevant and warranted.

CONCLUSION

Cells are continuously created and eliminated in a communal effort to optimize tissue function efficiency and overall health. Unsurprisingly then, unintentional malfunctions in these systems, whose consequences range from the undesired elimination of healthy cells to the undesired persistence of unwanted cells, manifest in various human pathologies. As research into the mechanisms that mediate autophagy and apoptosis generates an ever-expanding understanding of their complexity, it becomes increasingly relevant to consider these processes together. Importantly, as we identify new potential interventional targets through which to alter human biology, we must simultaneously be aware of the potential corresponding side effects such interventions may incur.

GRANTS

This work was supported in part by Natural Sciences and Engineering Research Council of Canada Grant RGPIN 258590 (J. Quadrilatero).

DISCLOSURES

No conflicts of interest, financial or otherwise, are declared by the authors.

AUTHOR CONTRIBUTIONS

D.B. prepared figures; D.B. and J.Q. drafted manuscript; D.B. and J.Q. edited and revised manuscript; D.B. and J.Q. approved final version of manuscript.

REFERENCES

  • 1. Alavian KN, Beutner G, Lazrove E, Sacchetti S, Park HA, Licznerski P, Li H, Nabili P, Hockensmith K, Graham M, Porter GA Jr, Jonas EA. An uncoupling channel within the c-subunit ring of the F1FO ATP synthase is the mitochondrial permeability transition pore. Proc Natl Acad Sci USA 111: 10580–10585, 2014. doi:10.1073/pnas.1401591111.
    Crossref | PubMed | ISI | Google Scholar
  • 2. Alers S, Löffler AS, Wesselborg S, Stork B. Role of AMPK-mTOR-Ulk1/2 in the regulation of autophagy: cross talk, shortcuts, and feedbacks. Mol Cell Biol 32: 2–11, 2012. doi:10.1128/MCB.06159-11.
    Crossref | PubMed | ISI | Google Scholar
  • 3. Alexander A, Cai SL, Kim J, Nanez A, Sahin M, MacLean KH, Inoki K, Guan KL, Shen J, Person MD, Kusewitt D, Mills GB, Kastan MB, Walker CL. ATM signals to TSC2 in the cytoplasm to regulate mTORC1 in response to ROS. Proc Natl Acad Sci USA 107: 4153–4158, 2010. [Erratum in Proc Natl Acad Sci USA 109: 8352, 2012.] doi:10.1073/pnas.0913860107.
    Crossref | PubMed | ISI | Google Scholar
  • 4. Aluvila S, Mandal T, Hustedt E, Fajer P, Choe JY, Oh KJ. Organization of the mitochondrial apoptotic BAK pore: oligomerization of the BAK homodimers. J Biol Chem 289: 2537–2551, 2014. doi:10.1074/jbc.M113.526806.
    Crossref | PubMed | ISI | Google Scholar
  • 5. Alway SE, Siu PM. Nuclear apoptosis contributes to sarcopenia. Exerc Sport Sci Rev 36: 51–57, 2008. doi:10.1097/JES.0b013e318168e9dc.
    Crossref | PubMed | ISI | Google Scholar
  • 6. Arena G, Gelmetti V, Torosantucci L, Vignone D, Lamorte G, De Rosa P, Cilia E, Jonas EA, Valente EM. PINK1 protects against cell death induced by mitochondrial depolarization, by phosphorylating Bcl-xL and impairing its pro-apoptotic cleavage. Cell Death Differ 20: 920–930, 2013. doi:10.1038/cdd.2013.19.
    Crossref | PubMed | ISI | Google Scholar
  • 7. Ashrafi G, Schwarz TL. The pathways of mitophagy for quality control and clearance of mitochondria. Cell Death Differ 20: 31–42, 2013. doi:10.1038/cdd.2012.81.
    Crossref | PubMed | ISI | Google Scholar
  • 8. Auré K, Fayet G, Leroy JP, Lacène E, Romero NB, Lombès A. Apoptosis in mitochondrial myopathies is linked to mitochondrial proliferation. Brain 129: 1249–1259, 2006. doi:10.1093/brain/awl061.
    Crossref | PubMed | ISI | Google Scholar
  • 9. Axe EL, Walker SA, Manifava M, Chandra P, Roderick HL, Habermann A, Griffiths G, Ktistakis NT. Autophagosome formation from membrane compartments enriched in phosphatidylinositol 3-phosphate and dynamically connected to the endoplasmic reticulum. J Cell Biol 182: 685–701, 2008. doi:10.1083/jcb.200803137.
    Crossref | PubMed | ISI | Google Scholar
  • 10. Baechler BL, Bloemberg D, Quadrilatero J. Mitophagy regulates mitochondrial network signaling, oxidative stress, and apoptosis during myoblast differentiation. Autophagy 1–14, 2019. doi:10.1080/15548627.2019.1591672.
    Crossref | PubMed | ISI | Google Scholar
  • 11. Baines CP, Kaiser RA, Sheiko T, Craigen WJ, Molkentin JD. Voltage-dependent anion channels are dispensable for mitochondrial-dependent cell death. Nat Cell Biol 9: 550–555, 2007. doi:10.1038/ncb1575.
    Crossref | PubMed | ISI | Google Scholar
  • 12. Batlevi Y, La Spada AR. Mitochondrial autophagy in neural function, neurodegenerative disease, neuron cell death, and aging. Neurobiol Dis 43: 46–51, 2011. doi:10.1016/j.nbd.2010.09.009.
    Crossref | PubMed | ISI | Google Scholar
  • 13. Bellot G, Garcia-Medina R, Gounon P, Chiche J, Roux D, Pouysségur J, Mazure NM. Hypoxia-induced autophagy is mediated through hypoxia-inducible factor induction of BNIP3 and BNIP3L via their BH3 domains. Mol Cell Biol 29: 2570–2581, 2009. doi:10.1128/MCB.00166-09.
    Crossref | PubMed | ISI | Google Scholar
  • 14. Bernardi P. The mitochondrial permeability transition pore: a mystery solved? Front Physiol 4: 95, 2013. doi:10.3389/fphys.2013.00095.
    Crossref | PubMed | ISI | Google Scholar
  • 15. Bernardi P, Krauskopf A, Basso E, Petronilli V, Blalchy-Dyson E, Di Lisa F, Forte MA. The mitochondrial permeability transition from in vitro artifact to disease target. FEBS J 273: 2077–2099, 2006. doi:10.1111/j.1742-4658.2006.05213.x.
    Crossref | PubMed | ISI | Google Scholar
  • 16. Bernardi P, Rasola A, Forte M, Lippe G. The mitochondrial permeability transition pore: channel formation by F-ATP synthase, integration in signal transduction, and role in pathophysiology. Physiol Rev 95: 1111–1155, 2015. doi:10.1152/physrev.00001.2015.
    Link | ISI | Google Scholar
  • 17. Betin VM, Lane JD. Caspase cleavage of Atg4D stimulates GABARAP-L1 processing and triggers mitochondrial targeting and apoptosis. J Cell Sci 122: 2554–2566, 2009. doi:10.1242/jcs.046250.
    Crossref | PubMed | ISI | Google Scholar
  • 18. Beutner G, Alavian KN, Jonas EA, Porter GA Jr. The mitochondrial permeability transition pore and ATP synthase. Handb Exp Pharmacol 240: 21–46, 2017. [Erratum in Handb Exp Pharmacol 240: 489, 2017.] doi:10.1007/164_2016_5.
    Crossref | PubMed | Google Scholar
  • 19. Beutner G, Rück A, Riede B, Brdiczka D. Complexes between porin, hexokinase, mitochondrial creatine kinase and adenylate translocator display properties of the permeability transition pore. Implication for regulation of permeability transition by the kinases. Biochim Biophys Acta 1368: 7–18, 1998. doi:10.1016/S0005-2736(97)00175-2.
    Crossref | PubMed | ISI | Google Scholar
  • 20. Bhujabal Z, Birgisdottir AB, Sjøttem E, Brenne HB, Øvervatn A, Habisov S, Kirkin V, Lamark T, Johansen T. FKBP8 recruits LC3A to mediate Parkin-independent mitophagy. EMBO Rep 18: 947–961, 2017. doi:10.15252/embr.201643147.
    Crossref | PubMed | ISI | Google Scholar
  • 21. Bjørkøy G, Lamark T, Brech A, Outzen H, Perander M, Overvatn A, Stenmark H, Johansen T. p62/SQSTM1 forms protein aggregates degraded by autophagy and has a protective effect on huntingtin-induced cell death. J Cell Biol 171: 603–614, 2005. doi:10.1083/jcb.200507002.
    Crossref | PubMed | ISI | Google Scholar
  • 22. Bleicken S, Jeschke G, Stegmueller C, Salvador-Gallego R, García-Sáez AJ, Bordignon E. Structural model of active Bax at the membrane. Mol Cell 56: 496–505, 2014. doi:10.1016/j.molcel.2014.09.022.
    Crossref | PubMed | ISI | Google Scholar
  • 23. Bloemberg D, Quadrilatero J. Effect of mitochondrial fission inhibition on C2C12 differentiation. Data Brief 7: 634–640, 2016. doi:10.1016/j.dib.2016.02.070.
    Crossref | PubMed | ISI | Google Scholar
  • 24. Bloemberg D, Quadrilatero J. Mitochondrial pro-apoptotic indices do not precede the transient caspase activation associated with myogenesis. Biochim Biophys Acta 1843: 2926–2936, 2014. doi:10.1016/j.bbamcr.2014.09.002.
    Crossref | PubMed | ISI | Google Scholar
  • 25. Boya P, González-Polo RA, Casares N, Perfettini JL, Dessen P, Larochette N, Métivier D, Meley D, Souquere S, Yoshimori T, Pierron G, Codogno P, Kroemer G. Inhibition of macroautophagy triggers apoptosis. Mol Cell Biol 25: 1025–1040, 2005. doi:10.1128/MCB.25.3.1025-1040.2005.
    Crossref | PubMed | ISI | Google Scholar
  • 26. Braun TP, Szumowski M, Levasseur PR, Grossberg AJ, Zhu X, Agarwal A, Marks DL. Muscle atrophy in response to cytotoxic chemotherapy is dependent on intact glucocorticoid signaling in skeletal muscle. PLoS One 9: e106489 , 2014. doi:10.1371/journal.pone.0106489.
    Crossref | PubMed | ISI | Google Scholar
  • 27. Bruusgaard JC, Gundersen K. In vivo time-lapse microscopy reveals no loss of murine myonuclei during weeks of muscle atrophy. J Clin Invest 118: 1450–1457, 2008. doi:10.1172/JCI34022.
    Crossref | PubMed | ISI | Google Scholar
  • 28. Bujak AL, Crane JD, Lally JS, Ford RJ, Kang SJ, Rebalka IA, Green AE, Kemp BE, Hawke TJ, Schertzer JD, Steinberg GR. AMPK activation of muscle autophagy prevents fasting-induced hypoglycemia and myopathy during aging. Cell Metab 21: 883–890, 2015. doi:10.1016/j.cmet.2015.05.016.
    Crossref | PubMed | ISI | Google Scholar
  • 29. Cagnol S, Chambard JC. ERK and cell death: mechanisms of ERK-induced cell death--apoptosis, autophagy and senescence. FEBS J 277: 2–21, 2010. doi:10.1111/j.1742-4658.2009.07366.x.
    Crossref | PubMed | ISI | Google Scholar
  • 30. Cao L, Xu J, Lin Y, Zhao X, Liu X, Chi Z. Autophagy is upregulated in rats with status epilepticus and partly inhibited by Vitamin E. Biochem Biophys Res Commun 379: 949–953, 2009. doi:10.1016/j.bbrc.2008.12.178.
    Crossref | PubMed | ISI | Google Scholar
  • 31. Carmignac V, Svensson M, Körner Z, Elowsson L, Matsumura C, Gawlik KI, Allamand V, Durbeej M. Autophagy is increased in laminin α2 chain-deficient muscle and its inhibition improves muscle morphology in a mouse model of MDC1A. Hum Mol Genet 20: 4891–4902, 2011. doi:10.1093/hmg/ddr427.
    Crossref | PubMed | ISI | Google Scholar
  • 32. Carra S, Seguin SJ, Lambert H, Landry J. HspB8 chaperone activity toward poly(Q)-containing proteins depends on its association with Bag3, a stimulator of macroautophagy. J Biol Chem 283: 1437–1444, 2008. doi:10.1074/jbc.M706304200.
    Crossref | PubMed | ISI | Google Scholar
  • 33. Castagnaro S, Pellegrini C, Pellegrini M, Chrisam M, Sabatelli P, Toni S, Grumati P, Ripamonti C, Pratelli L, Maraldi NM, Cocchi D, Righi V, Faldini C, Sandri M, Bonaldo P, Merlini L. Autophagy activation in COL6 myopathic patients by a low-protein-diet pilot trial. Autophagy 12: 2484–2495, 2016. doi:10.1080/15548627.2016.1231279.
    Crossref | PubMed | ISI | Google Scholar
  • 34. Castets P, Frank S, Sinnreich M, Rüegg MA. “Get the balance right”: pathological significance of autophagy perturbation in neuromuscular disorders. J Neuromuscul Dis 3: 127–155, 2016. doi:10.3233/JND-160153.
    Crossref | PubMed | Google Scholar
  • 35. Chaanine AH, Jeong D, Liang L, Chemaly ER, Fish K, Gordon RE, Hajjar RJ. JNK modulates FOXO3a for the expression of the mitochondrial death and mitophagy marker BNIP3 in pathological hypertrophy and in heart failure. Cell Death Dis 3: 265, 2012. doi:10.1038/cddis.2012.5.
    Crossref | PubMed | ISI | Google Scholar
  • 36. Chan NC, Salazar AM, Pham AH, Sweredoski MJ, Kolawa NJ, Graham RL, Hess S, Chan DC. Broad activation of the ubiquitin-proteasome system by Parkin is critical for mitophagy. Hum Mol Genet 20: 1726–1737, 2011. doi:10.1093/hmg/ddr048.
    Crossref | PubMed | ISI | Google Scholar
  • 37. Chen HC, Kanai M, Inoue-Yamauchi A, Tu HC, Huang Y, Ren D, Kim H, Takeda S, Reyna DE, Chan PM, Ganesan YT, Liao CP, Gavathiotis E, Hsieh JJ, Cheng EH. An interconnected hierarchical model of cell death regulation by the BCL-2 family. Nat Cell Biol 17: 1270–1281, 2015. doi:10.1038/ncb3236.
    Crossref | PubMed | ISI | Google Scholar
  • 38. Chen Y, McMillan-Ward E, Kong J, Israels SJ, Gibson SB. Oxidative stress induces autophagic cell death independent of apoptosis in transformed and cancer cells. Cell Death Differ 15: 171–182, 2008. doi:10.1038/sj.cdd.4402233.
    Crossref | PubMed | ISI | Google Scholar
  • 39. Choubey V, Cagalinec M, Liiv J, Safiulina D, Hickey MA, Kuum M, Liiv M, Anwar T, Eskelinen EL, Kaasik A. BECN1 is involved in the initiation of mitophagy: it facilitates PARK2 translocation to mitochondria. Autophagy 10: 1105–1119, 2014. doi:10.4161/auto.28615.
    Crossref | PubMed | ISI | Google Scholar
  • 40. Chrisam M, Pirozzi M, Castagnaro S, Blaauw B, Polishchuck R, Cecconi F, Grumati P, Bonaldo P. Reactivation of autophagy by spermidine ameliorates the myopathic defects of collagen VI-null mice. Autophagy 11: 2142–2152, 2015. doi:10.1080/15548627.2015.1108508.
    Crossref | PubMed | ISI | Google Scholar
  • 41. Chu CT, Ji J, Dagda RK, Jiang JF, Tyurina YY, Kapralov AA, Tyurin VA, Yanamala N, Shrivastava IH, Mohammadyani D, Wang KZQ, Zhu J, Klein-Seetharaman J, Balasubramanian K, Amoscato AA, Borisenko G, Huang Z, Gusdon AM, Cheikhi A, Steer EK, Wang R, Baty C, Watkins S, Bahar I, Bayir H, Kagan VE. Cardiolipin externalization to the outer mitochondrial membrane acts as an elimination signal for mitophagy in neuronal cells. Nat Cell Biol 15: 1197–1205, 2013. doi:10.1038/ncb2837.
    Crossref | PubMed | ISI | Google Scholar
  • 42. Cohen GM. Caspases: the executioners of apoptosis. Biochem J 326: 1–16, 1997. doi:10.1042/bj3260001.
    Crossref | PubMed | ISI | Google Scholar
  • 43. Costello MJ, Brennan LA, Basu S, Chauss D, Mohamed A, Gilliland KO, Johnsen S, Menko S, Kantorow M. Autophagy and mitophagy participate in ocular lens organelle degradation. Exp Eye Res 116: 141–150, 2013. doi:10.1016/j.exer.2013.08.017.
    Crossref | PubMed | ISI | Google Scholar
  • 44. Crighton D, Wilkinson S, O’Prey J, Syed N, Smith P, Harrison PR, Gasco M, Garrone O, Crook T, Ryan KM. DRAM, a p53-induced modulator of autophagy, is critical for apoptosis. Cell 126: 121–134, 2006. doi:10.1016/j.cell.2006.05.034.
    Crossref | PubMed | ISI | Google Scholar
  • 45. Cung TT, Morel O, Cayla G, Rioufol G, Garcia-Dorado D, Angoulvant D, Bonnefoy-Cudraz E, Guérin P, Elbaz M, Delarche N, Coste P, Vanzetto G, Metge M, Aupetit JF, Jouve B, Motreff P, Tron C, Labeque JN, Steg PG, Cottin Y, Range G, Clerc J, Claeys MJ, Coussement P, Prunier F, Moulin F, Roth O, Belle L, Dubois P, Barragan P, Gilard M, Piot C, Colin P, De Poli F, Morice MC, Ider O, Dubois-Randé JL, Unterseeh T, Le Breton H, Béard T, Blanchard D, Grollier G, Malquarti V, Staat P, Sudre A, Elmer E, Hansson MJ, Bergerot C, Boussaha I, Jossan C, Derumeaux G, Mewton N, Ovize M. Cyclosporine before PCI in patients with acute myocardial Infarction. N Engl J Med 373: 1021–1031, 2015. doi:10.1056/NEJMoa1505489.
    Crossref | PubMed | ISI | Google Scholar
  • 46. da Costa CA, Sunyach C, Giaime E, West A, Corti O, Brice A, Safe S, Abou-Sleiman PM, Wood NW, Takahashi H, Goldberg MS, Shen J, Checler F. Transcriptional repression of p53 by parkin and impairment by mutations associated with autosomal recessive juvenile Parkinson’s disease. Nat Cell Biol 11: 1370–1375, 2009. doi:10.1038/ncb1981.
    Crossref | PubMed | ISI | Google Scholar
  • 47. de Cabo R, Carmona-Gutierrez D, Bernier M, Hall MN, Madeo F. The search for antiaging interventions: from elixirs to fasting regimens. Cell 157: 1515–1526, 2014. doi:10.1016/j.cell.2014.05.031.
    Crossref | PubMed | ISI | Google Scholar
  • 48. De Palma C, Morisi F, Cheli S, Pambianco S, Cappello V, Vezzoli M, Rovere-Querini P, Moggio M, Ripolone M, Francolini M, Sandri M, Clementi E. Autophagy as a new therapeutic target in Duchenne muscular dystrophy. Cell Death Dis 3: e418 , 2012. doi:10.1038/cddis.2012.159.
    Crossref | PubMed | ISI | Google Scholar
  • 49. Deas E, Plun-Favreau H, Gandhi S, Desmond H, Kjaer S, Loh SH, Renton AE, Harvey RJ, Whitworth AJ, Martins LM, Abramov AY, Wood NW. PINK1 cleavage at position A103 by the mitochondrial protease PARL. Hum Mol Genet 20: 867–879, 2011. doi:10.1093/hmg/ddq526.
    Crossref | PubMed | ISI | Google Scholar
  • 50. Degenhardt K, Mathew R, Beaudoin B, Bray K, Anderson D, Chen G, Mukherjee C, Shi Y, Gélinas C, Fan Y, Nelson DA, Jin S, White E. Autophagy promotes tumor cell survival and restricts necrosis, inflammation, and tumorigenesis. Cancer Cell 10: 51–64, 2006. doi:10.1016/j.ccr.2006.06.001.
    Crossref | PubMed | ISI | Google Scholar
  • 51. Deveraux QL, Roy N, Stennicke HR, Van Arsdale T, Zhou Q, Srinivasula SM, Alnemri ES, Salvesen GS, Reed JC. IAPs block apoptotic events induced by caspase-8 and cytochrome c by direct inhibition of distinct caspases. EMBO J 17: 2215–2223, 1998. doi:10.1093/emboj/17.8.2215.
    Crossref | PubMed | ISI | Google Scholar
  • 52. Dhanasekaran DN, Reddy EP. JNK-signaling: A multiplexing hub in programmed cell death. Genes Cancer 8: 682–694, 2017. doi:10.18632/genesandcancer.155.
    Crossref | PubMed | Google Scholar
  • 53. Ding WX, Ni HM, Li M, Liao Y, Chen X, Stolz DB, Dorn GW II, Yin XM. Nix is critical to two distinct phases of mitophagy, reactive oxygen species-mediated autophagy induction and Parkin-ubiquitin-p62-mediated mitochondrial priming. J Biol Chem 285: 27879–27890, 2010. doi:10.1074/jbc.M110.119537.
    Crossref | PubMed | ISI | Google Scholar
  • 54. Dobrowolny G, Aucello M, Rizzuto E, Beccafico S, Mammucari C, Boncompagni S, Belia S, Wannenes F, Nicoletti C, Del Prete Z, Rosenthal N, Molinaro M, Protasi F, Fanò G, Sandri M, Musarò A. Skeletal muscle is a primary target of SOD1G93A-mediated toxicity. Cell Metab 8: 425–436, 2008. doi:10.1016/j.cmet.2008.09.002.
    Crossref | PubMed | ISI | Google Scholar
  • 55. Drummond MJ, Addison O, Brunker L, Hopkins PN, McClain DA, LaStayo PC, Marcus RL. Downregulation of E3 ubiquitin ligases and mitophagy-related genes in skeletal muscle of physically inactive, frail older women: a cross-sectional comparison. J Gerontol A Biol Sci Med Sci 69: 1040–1048, 2014. doi:10.1093/gerona/glu004.
    Crossref | PubMed | ISI | Google Scholar
  • 56. Du J, Wang X, Miereles C, Bailey JL, Debigare R, Zheng B, Price SR, Mitch WE. Activation of caspase-3 is an initial step triggering accelerated muscle proteolysis in catabolic conditions. J Clin Invest 113: 115–123, 2004. doi:10.1172/JCI18330.
    Crossref | PubMed | ISI | Google Scholar
  • 57. Dupont-Versteegden EE. Apoptosis in muscle atrophy: relevance to sarcopenia. Exp Gerontol 40: 473–481, 2005. doi:10.1016/j.exger.2005.04.003.
    Crossref | PubMed | ISI | Google Scholar
  • 58. Duran A, Linares JF, Galvez AS, Wikenheiser K, Flores JM, Diaz-Meco MT, Moscat J. The signaling adaptor p62 is an important NF-kappaB mediator in tumorigenesis. Cancer Cell 13: 343–354, 2008. doi:10.1016/j.ccr.2008.02.001.
    Crossref | PubMed | ISI | Google Scholar
  • 59. Elustondo PA, Nichols M, Negoda A, Thirumaran A, Zakharian E, Robertson GS, Pavlov EV. Mitochondrial permeability transition pore induction is linked to formation of the complex of ATPase C-subunit, polyhydroxybutyrate and inorganic polyphosphate. Cell Death Discov 2: 16070, 2016. doi:10.1038/cddiscovery.2016.70.
    Crossref | PubMed | ISI | Google Scholar
  • 60. Fan J, Yang X, Li J, Shu Z, Dai J, Liu X, Li B, Jia S, Kou X, Yang Y, Chen N. Spermidine coupled with exercise rescues skeletal muscle atrophy from D-gal-induced aging rats through enhanced autophagy and reduced apoptosis via AMPK-FOXO3a signal pathway. Oncotarget 8: 17475–17490, 2017. doi:10.18632/oncotarget.15728.
    Crossref | PubMed | Google Scholar
  • 61. Feng Z, Hu W, de Stanchina E, Teresky AK, Jin S, Lowe S, Levine AJ. The regulation of AMPK beta1, TSC2, and PTEN expression by p53: stress, cell and tissue specificity, and the role of these gene products in modulating the IGF-1-AKT-mTOR pathways. Cancer Res 67: 3043–3053, 2007. doi:10.1158/0008-5472.CAN-06-4149.
    Crossref | PubMed | ISI | Google Scholar
  • 62. Fernando P, Megeney LA. Is caspase-dependent apoptosis only cell differentiation taken to the extreme? FASEB J 21: 8–17, 2007. doi:10.1096/fj.06-5912hyp.
    Crossref | PubMed | ISI | Google Scholar
  • 63. Fimia GM, Stoykova A, Romagnoli A, Giunta L, Di Bartolomeo S, Nardacci R, Corazzari M, Fuoco C, Ucar A, Schwartz P, Gruss P, Piacentini M, Chowdhury K, Cecconi F. Ambra1 regulates autophagy and development of the nervous system. Nature 447: 1121–1125, 2007. doi:10.1038/nature05925.
    Crossref | PubMed | ISI | Google Scholar
  • 64. Fischer U, Jänicke RU, Schulze-Osthoff K. Many cuts to ruin: a comprehensive update of caspase substrates. Cell Death Differ 10: 76–100, 2003. doi:10.1038/sj.cdd.4401160.
    Crossref | PubMed | ISI | Google Scholar
  • 65. Frank M, Duvezin-Caubet S, Koob S, Occhipinti A, Jagasia R, Petcherski A, Ruonala MO, Priault M, Salin B, Reichert AS. Mitophagy is triggered by mild oxidative stress in a mitochondrial fission dependent manner. Biochim Biophys Acta 1823: 2297–2310, 2012. doi:10.1016/j.bbamcr.2012.08.007.
    Crossref | PubMed | ISI | Google Scholar
  • 66. Fry CS, Drummond MJ, Lujan HL, DiCarlo SE, Rasmussen BB. Paraplegia increases skeletal muscle autophagy. Muscle Nerve 46: 793–798, 2012. doi:10.1002/mus.23423.
    Crossref | PubMed | ISI | Google Scholar
  • 67. Furuya N, Ikeda S, Sato S, Soma S, Ezaki J, Oliva Trejo JA, Takeda-Ezaki M, Fujimura T, Arikawa-Hirasawa E, Tada N, Komatsu M, Tanaka K, Kominami E, Hattori N, Ueno T. PARK2/Parkin-mediated mitochondrial clearance contributes to proteasome activation during slow-twitch muscle atrophy via NFE2L1 nuclear translocation. Autophagy 10: 631–641, 2014. doi:10.4161/auto.27785.
    Crossref | PubMed | ISI | Google Scholar
  • 68. 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, 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, De Laurenzi V, De Maria R, Debatin KM, DeBerardinis RJ, Deshmukh M, Di Daniele N, Di Virgilio F, 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 JC, Martin SJ, Martinou JC, 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 HU, Sistigu A, Stockwell BR, Strasser A, Szabadkai G, Tait SWG, Tang D, Tavernarakis N, Thorburn A, Tsujimoto Y, Turk B, Vanden Berghe T, Vandenabeele P, Vander Heiden MG, 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. Molecular mechanisms of cell death: recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death Differ 25: 486–541, 2018. doi:10.1038/s41418-017-0012-4.
    Crossref | PubMed | ISI | Google Scholar
  • 69. Gamerdinger M, Kaya AM, Wolfrum U, Clement AM, Behl C. BAG3 mediates chaperone-based aggresome-targeting and selective autophagy of misfolded proteins. EMBO Rep 12: 149–156, 2011. doi:10.1038/embor.2010.203.
    Crossref | PubMed | ISI | Google Scholar
  • 70. Ganley IG, Lam H, Wang J, Ding X, Chen S, Jiang X. ULK1.ATG13.FIP200 complex mediates mTOR signaling and is essential for autophagy. J Biol Chem 284: 12297–12305, 2009. doi:10.1074/jbc.M900573200.
    Crossref | PubMed | ISI | Google Scholar
  • 71. Garofalo T, Matarrese P, Manganelli V, Marconi M, Tinari A, Gambardella L, Faggioni A, Misasi R, Sorice M, Malorni W. Evidence for the involvement of lipid rafts localized at the ER-mitochondria associated membranes in autophagosome formation. Autophagy 12: 917–935, 2016. doi:10.1080/15548627.2016.1160971.
    Crossref | PubMed | ISI | Google Scholar
  • 72. Gegg ME, Cooper JM, Chau KY, Rojo M, Schapira AH, Taanman JW. Mitofusin 1 and mitofusin 2 are ubiquitinated in a PINK1/parkin-dependent manner upon induction of mitophagy. Hum Mol Genet 19: 4861–4870, 2010. doi:10.1093/hmg/ddq419.
    Crossref | PubMed | ISI | Google Scholar
  • 73. Geisler S, Holmström KM, Skujat D, Fiesel FC, Rothfuss OC, Kahle PJ, Springer W. PINK1/Parkin-mediated mitophagy is dependent on VDAC1 and p62/SQSTM1. Nat Cell Biol 12: 119–131, 2010. doi:10.1038/ncb2012.
    Crossref | PubMed | ISI | Google Scholar
  • 74. Gelmetti V, De Rosa P, Torosantucci L, Marini ES, Romagnoli A, Di Rienzo M, Arena G, Vignone D, Fimia GM, Valente EM. PINK1 and BECN1 relocalize at mitochondria-associated membranes during mitophagy and promote ER-mitochondria tethering and autophagosome formation. Autophagy 13: 654–669, 2017. doi:10.1080/15548627.2016.1277309.
    Crossref | PubMed | ISI | Google Scholar
  • 75. Giorgio V, von Stockum S, Antoniel M, Fabbro A, Fogolari F, Forte M, Glick GD, Petronilli V, Zoratti M, Szabó I, Lippe G, Bernardi P. Dimers of mitochondrial ATP synthase form the permeability transition pore. Proc Natl Acad Sci USA 110: 5887–5892, 2013. doi:10.1073/pnas.1217823110.
    Crossref | PubMed | ISI | Google Scholar
  • 76. Gomes LC, Di Benedetto G, Scorrano L. During autophagy mitochondria elongate, are spared from degradation and sustain cell viability. Nat Cell Biol 13: 589–598, 2011. doi:10.1038/ncb2220.
    Crossref | PubMed | ISI | Google Scholar
  • 77. Gomez L, Thibault H, Gharib A, Dumont JM, Vuagniaux G, Scalfaro P, Derumeaux G, Ovize M. Inhibition of mitochondrial permeability transition improves functional recovery and reduces mortality following acute myocardial infarction in mice. Am J Physiol Heart Circ Physiol 293: H1654–H1661, 2007. doi:10.1152/ajpheart.01378.2006.
    Link | ISI | Google Scholar
  • 78. Gouspillou G, Godin R, Piquereau J, Picard M, Mofarrahi M, Mathew J, Purves-Smith FM, Sgarioto N, Hepple RT, Burelle Y, Hussain SNA. Protective role of Parkin in skeletal muscle contractile and mitochondrial function. J Physiol 596: 2565–2579, 2018. doi:10.1113/JP275604.
    Crossref | PubMed | ISI | Google Scholar
  • 79. Grootjans S, Vanden Berghe T, Vandenabeele P. Initiation and execution mechanisms of necroptosis: an overview. Cell Death Differ 24: 1184–1195, 2017. doi:10.1038/cdd.2017.65.
    Crossref | PubMed | ISI | Google Scholar
  • 80. Große L, Wurm CA, Brüser C, Neumann D, Jans DC, Jakobs S. Bax assembles into large ring-like structures remodeling the mitochondrial outer membrane in apoptosis. EMBO J 35: 402–413, 2016. doi:10.15252/embj.201592789.
    Crossref | PubMed | ISI | Google Scholar
  • 81. Grumati P, Coletto L, Sabatelli P, Cescon M, Angelin A, Bertaggia E, Blaauw B, Urciuolo A, Tiepolo T, Merlini L, Maraldi NM, Bernardi P, Sandri M, Bonaldo P. Autophagy is defective in collagen VI muscular dystrophies, and its reactivation rescues myofiber degeneration. Nat Med 16: 1313–1320, 2010. doi:10.1038/nm.2247.
    Crossref | PubMed | ISI | Google Scholar
  • 82. Guicciardi ME, Gores GJ. Life and death by death receptors. FASEB J 23: 1625–1637, 2009. doi:10.1096/fj.08-111005.
    Crossref | PubMed | ISI | Google Scholar
  • 83. Guo JY, Karsli-Uzunbas G, Mathew R, Aisner SC, Kamphorst JJ, Strohecker AM, Chen G, Price S, Lu W, Teng X, Snyder E, Santanam U, Dipaola RS, Jacks T, Rabinowitz JD, White E. Autophagy suppresses progression of K-ras-induced lung tumors to oncocytomas and maintains lipid homeostasis. Genes Dev 27: 1447–1461, 2013. doi:10.1101/gad.219642.113.
    Crossref | PubMed | ISI | Google Scholar
  • 84. Guo Y, Meng J, Tang Y, Wang T, Wei B, Feng R, Gong B, Wang H, Ji G, Lu Z. AMP-activated kinase α2 deficiency protects mice from denervation-induced skeletal muscle atrophy. Arch Biochem Biophys 600: 56–60, 2016. doi:10.1016/j.abb.2016.04.015.
    Crossref | PubMed | ISI | Google Scholar
  • 85. Gustafsson AB, Gottlieb RA. Bcl-2 family members and apoptosis, taken to heart. Am J Physiol Cell Physiol 292: C45–C51, 2007. doi:10.1152/ajpcell.00229.2006.
    Link | ISI | Google Scholar
  • 86. Gwinn DM, Shackelford DB, Egan DF, Mihaylova MM, Mery A, Vasquez DS, Turk BE, Shaw RJ. AMPK phosphorylation of raptor mediates a metabolic checkpoint. Mol Cell 30: 214–226, 2008. doi:10.1016/j.molcel.2008.03.003.
    Crossref | PubMed | ISI | Google Scholar
  • 87. Hagenbuchner J, Lungkofler L, Kiechl-Kohlendorfer U, Viola G, Ferlin MG, Ausserlechner MJ, Obexer P. The tubulin inhibitor MG-2477 induces autophagy-regulated cell death, ROS accumulation and activation of FOXO3 in neuroblastoma. Oncotarget 8: 32009–32026, 2017. doi:10.18632/oncotarget.16434.
    Crossref | PubMed | Google Scholar
  • 88. Hailey DW, Rambold AS, Satpute-Krishnan P, Mitra K, Sougrat R, Kim PK, Lippincott-Schwartz J. Mitochondria supply membranes for autophagosome biogenesis during starvation. Cell 141: 656–667, 2010. doi:10.1016/j.cell.2010.04.009.
    Crossref | PubMed | ISI | Google Scholar
  • 89. Halestrap AP, Brenner C. The adenine nucleotide translocase: a central component of the mitochondrial permeability transition pore and key player in cell death. Curr Med Chem 10: 1507–1525, 2003. doi:10.2174/0929867033457278.
    Crossref | PubMed | ISI | Google Scholar
  • 90. Halling JF, Ringholm S, Olesen J, Prats C, Pilegaard H. Exercise training protects against aging-induced mitochondrial fragmentation in mouse skeletal muscle in a PGC-1α dependent manner. Exp Gerontol 96: 1–6, 2017. doi:10.1016/j.exger.2017.05.020.
    Crossref | PubMed | ISI | Google Scholar
  • 91. Hamacher-Brady A, Brady NR, Logue SE, Sayen MR, Jinno M, Kirshenbaum LA, Gottlieb RA, Gustafsson AB. Response to myocardial ischemia/reperfusion injury involves Bnip3 and autophagy. Cell Death Differ 14: 146–157, 2007. doi:10.1038/sj.cdd.4401936.
    Crossref | PubMed | ISI | Google Scholar
  • 92. Hamasaki M, Furuta N, Matsuda A, Nezu A, Yamamoto A, Fujita N, Oomori H, Noda T, Haraguchi T, Hiraoka Y, Amano A, Yoshimori T. Autophagosomes form at ER-mitochondria contact sites. Nature 495: 389–393, 2013. doi:10.1038/nature11910.
    Crossref | PubMed | ISI | Google Scholar
  • 93. Hanada T, Noda NN, Satomi Y, Ichimura Y, Fujioka Y, Takao T, Inagaki F, Ohsumi Y. The Atg12-Atg5 conjugate has a novel E3-like activity for protein lipidation in autophagy. J Biol Chem 282: 37298–37302, 2007. doi:10.1074/jbc.C700195200.
    Crossref | PubMed | ISI | Google Scholar
  • 94. Hanna RA, Quinsay MN, Orogo AM, Giang K, Rikka S, Gustafsson AB. Microtubule-associated protein 1 light chain 3 (LC3) interacts with Bnip3 protein to selectively remove endoplasmic reticulum and mitochondria via autophagy. J Biol Chem 287: 19094–19104, 2012. doi:10.1074/jbc.M111.322933.
    Crossref | PubMed | ISI | Google Scholar
  • 95. Hayashi-Nishino M, Fujita N, Noda T, Yamaguchi A, Yoshimori T, Yamamoto A. Electron tomography reveals the endoplasmic reticulum as a membrane source for autophagosome formation. Autophagy 6: 301–303, 2010. doi:10.4161/auto.6.2.11134.
    Crossref | PubMed | ISI | Google Scholar
  • 96. Hayashi-Nishino M, Fujita N, Noda T, Yamaguchi A, Yoshimori T, Yamamoto A. A subdomain of the endoplasmic reticulum forms a cradle for autophagosome formation. Nat Cell Biol 11: 1433–1437, 2009. doi:10.1038/ncb1991.
    Crossref | PubMed | ISI | Google Scholar
  • 97. He C, Bassik MC, Moresi V, Sun K, Wei Y, Zou Z, An Z, Loh J, Fisher J, Sun Q, Korsmeyer S, Packer M, May HI, Hill JA, Virgin HW, Gilpin C, Xiao G, Bassel-Duby R, Scherer PE, Levine B. Exercise-induced BCL2-regulated autophagy is required for muscle glucose homeostasis. Nature 481: 511–515, 2012. [Erratum in Nature 503: 146, 2013.] doi:10.1038/nature10758.
    Crossref | PubMed | ISI | Google Scholar
  • 98. He C, Klionsky DJ. Regulation mechanisms and signaling pathways of autophagy. Annu Rev Genet 43: 67–93, 2009. doi:10.1146/annurev-genet-102808-114910.
    Crossref | PubMed | ISI | Google Scholar
  • 99. He J, Carroll J, Ding S, Fearnley IM, Walker JE. Permeability transition in human mitochondria persists in the absence of peripheral stalk subunits of ATP synthase. Proc Natl Acad Sci USA 114: 9086–9091, 2017. doi:10.1073/pnas.1711201114.
    Crossref | PubMed | ISI | Google Scholar
  • 100. He J, Ford HC, Carroll J, Ding S, Fearnley IM, Walker JE. Persistence of the mitochondrial permeability transition in the absence of subunit c of human ATP synthase. Proc Natl Acad Sci USA 114: 3409–3414, 2017. doi:10.1073/pnas.1702357114.
    Crossref | PubMed | ISI | Google Scholar
  • 101. Hemelaar J, Lelyveld VS, Kessler BM, Ploegh HL. A single protease, Apg4B, is specific for the autophagy-related ubiquitin-like proteins GATE-16, MAP1-LC3, GABARAP, and Apg8L. J Biol Chem 278: 51841–51850, 2003. doi:10.1074/jbc.M308762200.
    Crossref | PubMed | ISI | Google Scholar
  • 102. Heo JM, Ordureau A, Paulo JA, Rinehart J, Harper JW. The PINK1-PARKIN mitochondrial ubiquitylation pathway drives a program of OPTN/NDP52 recruitment and TBK1 activation to promote mitophagy. Mol Cell 60: 7–20, 2015. doi:10.1016/j.molcel.2015.08.016.
    Crossref | PubMed | ISI | Google Scholar
  • 103. Higgins GC, Coughlan MT. Mitochondrial dysfunction and mitophagy: the beginning and end to diabetic nephropathy? Br J Pharmacol 171: 1917–1942, 2014. doi:10.1111/bph.12503.
    Crossref | PubMed | ISI | Google Scholar
  • 104. Hoshino A, Ariyoshi M, Okawa Y, Kaimoto S, Uchihashi M, Fukai K, Iwai-Kanai E, Ikeda K, Ueyama T, Ogata T, Matoba S. Inhibition of p53 preserves Parkin-mediated mitophagy and pancreatic β-cell function in diabetes. Proc Natl Acad Sci USA 111: 3116–3121, 2014. doi:10.1073/pnas.1318951111.
    Crossref | PubMed | ISI | Google Scholar
  • 105. Hoshino A, Matoba S, Iwai-Kanai E, Nakamura H, Kimata M, Nakaoka M, Katamura M, Okawa Y, Ariyoshi M, Mita Y, Ikeda K, Ueyama T, Okigaki M, Matsubara H. p53-TIGAR axis attenuates mitophagy to exacerbate cardiac damage after ischemia. J Mol Cell Cardiol 52: 175–184, 2012. doi:10.1016/j.yjmcc.2011.10.008.
    Crossref | PubMed | ISI | Google Scholar
  • 106. Hoshino A, Mita Y, Okawa Y, Ariyoshi M, Iwai-Kanai E, Ueyama T, Ikeda K, Ogata T, Matoba S. Cytosolic p53 inhibits Parkin-mediated mitophagy and promotes mitochondrial dysfunction in the mouse heart. Nat Commun 4: 2308, 2013. doi:10.1038/ncomms3308.
    Crossref | PubMed | ISI | Google Scholar
  • 107. Hosokawa N, Sasaki T, Iemura S, Natsume T, Hara T, Mizushima N. Atg101, a novel mammalian autophagy protein interacting with Atg13. Autophagy 5: 973–979, 2009. doi:10.4161/auto.5.7.9296.
    Crossref | PubMed | ISI | Google Scholar
  • 108. Hussain SN, Mofarrahi M, Sigala I, Kim HC, Vassilakopoulos T, Maltais F, Bellenis I, Chaturvedi R, Gottfried SB, Metrakos P, Danialou G, Matecki S, Jaber S, Petrof BJ, Goldberg P. Mechanical ventilation-induced diaphragm disuse in humans triggers autophagy. Am J Respir Crit Care Med 182: 1377–1386, 2010. doi:10.1164/rccm.201002-0234OC.
    Crossref | PubMed | ISI | Google Scholar
  • 109. Ichimura Y, Kumanomidou T, Sou YS, Mizushima T, Ezaki J, Ueno T, Kominami E, Yamane T, Tanaka K, Komatsu M. Structural basis for sorting mechanism of p62 in selective autophagy. J Biol Chem 283: 22847–22857, 2008. doi:10.1074/jbc.M802182200.
    Crossref | PubMed | ISI | Google Scholar
  • 110. Ikeda G, Matoba T, Nakano Y, Nagaoka K, Ishikita A, Nakano K, Funamoto D, Sunagawa K, Egashira K. Nanoparticle-mediated targeting of cyclosporine A enhances cardioprotection against ischemia-reperfusion injury through inhibition of mitochondrial permeability transition pore opening. Sci Rep 6: 20467, 2016. doi:10.1038/srep20467.
    Crossref | PubMed | ISI | Google Scholar
  • 111. Inoki K, Zhu T, Guan KL. TSC2 mediates cellular energy response to control cell growth and survival. Cell 115: 577–590, 2003. doi:10.1016/S0092-8674(03)00929-2.
    Crossref | PubMed | ISI | Google Scholar
  • 112. Irwin WA, Bergamin N, Sabatelli P, Reggiani C, Megighian A, Merlini L, Braghetta P, Columbaro M, Volpin D, Bressan GM, Bernardi P, Bonaldo P. Mitochondrial dysfunction and apoptosis in myopathic mice with collagen VI deficiency. Nat Genet 35: 367–371, 2003. doi:10.1038/ng1270.
    Crossref | PubMed | ISI | Google Scholar
  • 113. Izzo V, Bravo-San Pedro JM, Sica V, Kroemer G, Galluzzi L. Mitochondrial permeability transition: new findings and persisting uncertainties. Trends Cell Biol 26: 655–667, 2016. doi:10.1016/j.tcb.2016.04.006.
    Crossref | PubMed | ISI | Google Scholar
  • 114. Jamart C, Gomes AV, Dewey S, Deldicque L, Raymackers JM, Francaux M. Regulation of ubiquitin-proteasome and autophagy pathways after acute LPS and epoxomicin administration in mice. BMC Musculoskelet Disord 15: 166, 2014. doi:10.1186/1471-2474-15-166.
    Crossref | PubMed | ISI | Google Scholar
  • 115. Jeong HS, Choi HY, Lee ER, Kim JH, Jeon K, Lee HJ, Cho SG. Involvement of caspase-9 in autophagy-mediated cell survival pathway. Biochim Biophys Acta 1813: 80–90, 2011. doi:10.1016/j.bbamcr.2010.09.016.
    Crossref | PubMed | ISI | Google Scholar
  • 116. Jiao J, Demontis F. Skeletal muscle autophagy and its role in sarcopenia and organismal aging. Curr Opin Pharmacol 34: 1–6, 2017. doi:10.1016/j.coph.2017.03.009.
    Crossref | PubMed | ISI | Google Scholar
  • 117. Jin SM, Lazarou M, Wang C, Kane LA, Narendra DP, Youle RJ. Mitochondrial membrane potential regulates PINK1 import and proteolytic destabilization by PARL. J Cell Biol 191: 933–942, 2010. doi:10.1083/jcb.201008084.
    Crossref | PubMed | ISI | Google Scholar
  • 118. Joseph AM, Adhihetty PJ, Wawrzyniak NR, Wohlgemuth SE, Picca A, Kujoth GC, Prolla TA, Leeuwenburgh C. Dysregulation of mitochondrial quality control processes contribute to sarcopenia in a mouse model of premature aging. PLoS One 8: e69327 , 2013. doi:10.1371/journal.pone.0069327.
    Crossref | PubMed | ISI | Google Scholar
  • 119. Jung CH, Jun CB, Ro SH, Kim YM, Otto NM, Cao J, Kundu M, Kim DH. ULK-Atg13-FIP200 complexes mediate mTOR signaling to the autophagy machinery. Mol Biol Cell 20: 1992–2003, 2009. doi:10.1091/mbc.e08-12-1249.
    Crossref | PubMed | ISI | Google Scholar
  • 120. Jung CH, Ro SH, Cao J, Otto NM, Kim DH. mTOR regulation of autophagy. FEBS Lett 584: 1287–1295, 2010. doi:10.1016/j.febslet.2010.01.017.
    Crossref | PubMed | ISI | Google Scholar
  • 121. Kabeya Y, Mizushima N, Ueno T, Yamamoto A, Kirisako T, Noda T, Kominami E, Ohsumi Y, Yoshimori T. LC3, a mammalian homologue of yeast Apg8p, is localized in autophagosome membranes after processing. EMBO J 19: 5720–5728, 2000. doi:10.1093/emboj/19.21.5720.
    Crossref | PubMed | ISI | Google Scholar
  • 122. Kahns S, Kalai M, Jakobsen LD, Clark BF, Vandenabeele P, Jensen PH. Caspase-1 and caspase-8 cleave and inactivate cellular parkin. J Biol Chem 278: 23376–23380, 2003. doi:10.1074/jbc.M300495200.
    Crossref | PubMed | ISI | Google Scholar
  • 123. Kang C, Yeo D, Ji LL. Muscle immobilization activates mitophagy and disrupts mitochondrial dynamics in mice. Acta Physiol (Oxf) 218: 188–197, 2016. doi:10.1111/apha.12690.
    Crossref | PubMed | ISI | Google Scholar
  • 124. Kim E, Goraksha-Hicks P, Li L, Neufeld TP, Guan KL. Regulation of TORC1 by Rag GTPases in nutrient response. Nat Cell Biol 10: 935–945, 2008. doi:10.1038/ncb1753.
    Crossref | PubMed | ISI | Google Scholar
  • 125. Kim H, Rafiuddin-Shah M, Tu HC, Jeffers JR, Zambetti GP, Hsieh JJ, Cheng EH. Hierarchical regulation of mitochondrion-dependent apoptosis by BCL-2 subfamilies. Nat Cell Biol 8: 1348–1358, 2006. doi:10.1038/ncb1499.
    Crossref | PubMed | ISI | Google Scholar
  • 126. Kim H, Tu HC, Ren D, Takeuchi O, Jeffers JR, Zambetti GP, Hsieh JJ, Cheng EH. Stepwise activation of BAX and BAK by tBID, BIM, and PUMA initiates mitochondrial apoptosis. Mol Cell 36: 487–499, 2009. doi:10.1016/j.molcel.2009.09.030.
    Crossref | PubMed | ISI | Google Scholar
  • 127. Kim I, Rodriguez-Enriquez S, Lemasters JJ. Selective degradation of mitochondria by mitophagy. Arch Biochem Biophys 462: 245–253, 2007. doi:10.1016/j.abb.2007.03.034.
    Crossref | PubMed | ISI | Google Scholar
  • 128. Kim J, Kundu M, Viollet B, Guan KL. AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1. Nat Cell Biol 13: 132–141, 2011. doi:10.1038/ncb2152.
    Crossref | PubMed | ISI | Google Scholar
  • 129. Kim Y, Park J, Kim S, Song S, Kwon SK, Lee SH, Kitada T, Kim JM, Chung J. PINK1 controls mitochondrial localization of Parkin through direct phosphorylation. Biochem Biophys Res Commun 377: 975–980, 2008. doi:10.1016/j.bbrc.2008.10.104.
    Crossref | PubMed | ISI | Google Scholar
  • 130. Kirkin V, Lamark T, Sou YS, Bjørkøy G, Nunn JL, Bruun JA, Shvets E, McEwan DG, Clausen TH, Wild P, Bilusic I, Theurillat JP, Øvervatn A, Ishii T, Elazar Z, Komatsu M, Dikic I, Johansen T. A role for NBR1 in autophagosomal degradation of ubiquitinated substrates. Mol Cell 33: 505–516, 2009. doi:10.1016/j.molcel.2009.01.020.
    Crossref | PubMed | ISI | Google Scholar
  • 131. Kischkel FC, Hellbardt S, Behrmann I, Germer M, Pawlita M, Krammer PH, Peter ME. Cytotoxicity-dependent APO-1 (Fas/CD95)-associated proteins form a death-inducing signaling complex (DISC) with the receptor. EMBO J 14: 5579–5588, 1995. doi:10.1002/j.1460-2075.1995.tb00245.x.
    Crossref | PubMed | ISI | Google Scholar
  • 132. Kitada T, Asakawa S, Hattori N, Matsumine H, Yamamura Y, Minoshima S, Yokochi M, Mizuno Y, Shimizu N. Mutations in the parkin gene cause autosomal recessive juvenile parkinsonism. Nature 392: 605–608, 1998. doi:10.1038/33416.
    Crossref | PubMed | ISI | Google Scholar
  • 133. Klionsky DJ, Abdelmohsen K, Abe A, Abedin MJ, Abeliovich H, Acevedo Arozena A, , et al.. Guidelines for the use and interpretation of assays for monitoring autophagy (3rd edition). Autophagy 12: 1–222, 2016. [Erratum in Autophagy 12: 443, 2016.] doi:10.1080/15548627.2015.1100356.
    Crossref | PubMed | ISI | Google Scholar
  • 134. Kokoszka JE, Waymire KG, Levy SE, Sligh JE, Cai J, Jones DP, MacGregor GR, Wallace DC. The ADP/ATP translocator is not essential for the mitochondrial permeability transition pore. Nature 427: 461–465, 2004. doi:10.1038/nature02229.
    Crossref | PubMed | ISI | Google Scholar
  • 135. Komatsu M, Waguri S, Ueno T, Iwata J, Murata S, Tanida I, Ezaki J, Mizushima N, Ohsumi Y, Uchiyama Y, Kominami E, Tanaka K, Chiba T. Impairment of starvation-induced and constitutive autophagy in Atg7-deficient mice. J Cell Biol 169: 425–434, 2005. doi:10.1083/jcb.200412022.
    Crossref | PubMed | ISI | Google Scholar
  • 136. Kozakowska M, Pietraszek-Gremplewicz K, Jozkowicz A, Dulak J. The role of oxidative stress in skeletal muscle injury and regeneration: focus on antioxidant enzymes. J Muscle Res Cell Motil 36: 377–393, 2015. doi:10.1007/s10974-015-9438-9.
    Crossref | PubMed | ISI | Google Scholar
  • 137. Kroemer G. Autophagy: a druggable process that is deregulated in aging and human disease. J Clin Invest 125: 1–4, 2015. doi:10.1172/JCI78652.
    Crossref | PubMed | ISI | Google Scholar
  • 138. Kroemer G, Galluzzi L, Brenner C. Mitochondrial membrane permeabilization in cell death. Physiol Rev 87: 99–163, 2007. doi:10.1152/physrev.00013.2006.
    Link | ISI | Google Scholar
  • 139. Kubli DA, Cortez MQ, Moyzis AG, Najor RH, Lee Y, Gustafsson AB. PINK1 is dispensable for mitochondrial recruitment of parkin and activation of mitophagy in cardiac myocytes. PLoS One 10: e0130707 , 2015. doi:10.1371/journal.pone.0130707.
    Crossref | PubMed | ISI | Google Scholar
  • 140. Kubli DA, Zhang X, Lee Y, Hanna RA, Quinsay MN, Nguyen CK, Jimenez R, Petrosyan S, Murphy AN, Gustafsson AB. Parkin protein deficiency exacerbates cardiac injury and reduces survival following myocardial infarction. J Biol Chem 288: 915–926, 2013. doi:10.1074/jbc.M112.411363.
    Crossref | PubMed | ISI | Google Scholar
  • 141. Kuma A, Hatano M, Matsui M, Yamamoto A, Nakaya H, Yoshimori T, Ohsumi Y, Tokuhisa T, Mizushima N. The role of autophagy during the early neonatal starvation period. Nature 432: 1032–1036, 2004. doi:10.1038/nature03029.
    Crossref | PubMed | ISI | Google Scholar
  • 142. Kurihara Y, Kanki T, Aoki Y, Hirota Y, Saigusa T, Uchiumi T, Kang D. Mitophagy plays an essential role in reducing mitochondrial production of reactive oxygen species and mutation of mitochondrial DNA by maintaining mitochondrial quantity and quality in yeast. J Biol Chem 287: 3265–3272, 2012. doi:10.1074/jbc.M111.280156.
    Crossref | PubMed | ISI | Google Scholar
  • 143. 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 17: 525–535, 2005. doi:10.1016/j.molcel.2005.02.003.
    Crossref | PubMed | ISI | Google Scholar
  • 144. Lampert MA, Orogo AM, Najor RH, Hammerling BC, Leon LJ, Wang BJ, Kim T, Sussman MA, Gustafsson AB. BNIP3L/NIX and FUNDC1-mediated mitophagy is required for mitochondrial network remodeling during cardiac progenitor cell differentiation. Autophagy: 1–17, 2019. doi:10.1080/15548627.2019.1580095.
    Crossref | PubMed | ISI | Google Scholar
  • 145. Larsen BD, Rampalli S, Burns LE, Brunette S, Dilworth FJ, Megeney LA. Caspase 3/caspase-activated DNase promote cell differentiation by inducing DNA strand breaks. Proc Natl Acad Sci USA 107: 4230–4235, 2010. doi:10.1073/pnas.0913089107.
    Crossref | PubMed | ISI | Google Scholar
  • 146. Lazarou M, Jin SM, Kane LA, Youle RJ. Role of PINK1 binding to the TOM complex and alternate intracellular membranes in recruitment and activation of the E3 ligase Parkin. Dev Cell 22: 320–333, 2012. doi:10.1016/j.devcel.2011.12.014.
    Crossref | PubMed | ISI | Google Scholar
  • 147. Lazarou M, Sliter DA, Kane LA, Sarraf SA, Wang C, Burman JL, Sideris DP, Fogel AI, Youle RJ. The ubiquitin kinase PINK1 recruits autophagy receptors to induce mitophagy. Nature 524: 309–314, 2015. doi:10.1038/nature14893.
    Crossref | PubMed | ISI | Google Scholar
  • 148. Leduc-Gaudet JP, Reynaud O, Hussain SN, Gouspillou G. Parkin overexpression protects from ageing-related loss of muscle mass and strength. J Physiol 597: 1975–1991, 2019. doi:10.1113/JP277157.
    Crossref | PubMed | ISI | Google Scholar
  • 149. Lee JW, Park S, Takahashi Y, Wang HG. The association of AMPK with ULK1 regulates autophagy. PLoS One 5: e15394 , 2010. doi:10.1371/journal.pone.0015394.
    Crossref | PubMed | ISI | Google Scholar
  • 150. Lee Y, Lee HY, Hanna RA, Gustafsson AB. Mitochondrial autophagy by Bnip3 involves Drp1-mediated mitochondrial fission and recruitment of Parkin in cardiac myocytes. Am J Physiol Heart Circ Physiol 301: H1924–H1931, 2011. doi:10.1152/ajpheart.00368.2011.
    Link | ISI | Google Scholar
  • 151. 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 2: 183–192, 2002. doi:10.1016/S1535-6108(02)00127-7.
    Crossref | PubMed | ISI | Google Scholar
  • 152. Levine B, Kroemer G. Autophagy in the pathogenesis of disease. Cell 132: 27–42, 2008. doi:10.1016/j.cell.2007.12.018.
    Crossref | PubMed | ISI | Google Scholar
  • 153. Levine B, Packer M, Codogno P. Development of autophagy inducers in clinical medicine. J Clin Invest 125: 14–24, 2015. doi:10.1172/JCI73938.
    Crossref | PubMed | ISI | Google Scholar
  • 154. Li LY, Luo X, Wang X. Endonuclease G is an apoptotic DNase when released from mitochondria. Nature 412: 95–99, 2001. doi:10.1038/35083620.
    Crossref | PubMed | ISI | Google Scholar
  • 155. 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 91: 479–489, 1997. doi:10.1016/S0092-8674(00)80434-1.
    Crossref | PubMed | ISI | Google Scholar
  • 156. Li Q, Zhang T, Wang J, Zhang Z, Zhai Y, Yang GY, Sun X. Rapamycin attenuates mitochondrial dysfunction via activation of mitophagy in experimental ischemic stroke. Biochem Biophys Res Commun 444: 182–188, 2014. doi:10.1016/j.bbrc.2014.01.032.
    Crossref | PubMed | ISI | Google Scholar
  • 157. Li Y, Jiang J, Liu W, Wang H, Zhao L, Liu S, Li P, Zhang S, Sun C, Wu Y, Yu S, Li X, Zhang H, Qian H, Zhang D, Guo F, Zhai Q, Ding Q, Wang L, Ying H. microRNA-378 promotes autophagy and inhibits apoptosis in skeletal muscle. Proc Natl Acad Sci USA 115: E10849–E10858, 2018. doi:10.1073/pnas.1803377115.
    Crossref | PubMed | ISI | Google Scholar
  • 158. Liang XH, Jackson S, Seaman M, Brown K, Kempkes B, Hibshoosh H, Levine B. Induction of autophagy and inhibition of tumorigenesis by beclin 1. Nature 402: 672–676, 1999. doi:10.1038/45257.
    Crossref | PubMed | ISI | Google Scholar
  • 159. Lim JA, Li L, Kakhlon O, Myerowitz R, Raben N. Defects in calcium homeostasis and mitochondria can be reversed in Pompe disease. Autophagy 11: 385–402, 2015. doi:10.1080/15548627.2015.1009779.
    Crossref | PubMed | ISI | Google Scholar
  • 160. Lim SY, Hausenloy DJ, Arjun S, Price AN, Davidson SM, Lythgoe MF, Yellon DM. Mitochondrial cyclophilin-D as a potential therapeutic target for post-myocardial infarction heart failure. J Cell Mol Med 15: 2443–2451, 2011. doi:10.1111/j.1582-4934.2010.01235.x.
    Crossref | PubMed | ISI | Google Scholar
  • 161. Linkermann A, Konstantinidis K, Kitsis RN. Catch me if you can: targeting the mitochondrial permeability transition pore in myocardial infarction. Cell Death Differ 23: 1–2, 2016. doi:10.1038/cdd.2015.151.
    Crossref | PubMed | ISI | Google Scholar
  • 162. Lira VA, Okutsu M, Zhang M, Greene NP, Laker RC, Breen DS, Hoehn KL, Yan Z. Autophagy is required for exercise training-induced skeletal muscle adaptation and improvement of physical performance. FASEB J 27: 4184–4193, 2013. doi:10.1096/fj.13-228486.
    Crossref | PubMed | ISI | Google Scholar
  • 163. Liu B, Cheng Y, Zhang B, Bian HJ, Bao JK. Polygonatum cyrtonema lectin induces apoptosis and autophagy in human melanoma A375 cells through a mitochondria-mediated ROS-p38-p53 pathway. Cancer Lett 275: 54–60, 2009. doi:10.1016/j.canlet.2008.09.042.
    Crossref | PubMed | ISI | Google Scholar
  • 164. Liu L, Feng D, Chen G, Chen M, Zheng Q, Song P, Ma Q, Zhu C, Wang R, Qi W, Huang L, Xue P, Li B, Wang X, Jin H, Wang J, Yang F, Liu P, Zhu Y, Sui S, Chen Q. Mitochondrial outer-membrane protein FUNDC1 mediates hypoxia-induced mitophagy in mammalian cells. Nat Cell Biol 14: 177–185, 2012. doi:10.1038/ncb2422.
    Crossref | PubMed | ISI | Google Scholar
  • 166. Lu T, Gu M, Zhao Y, Zheng X, Xing C. Autophagy contributes to falcarindiol-induced cell death in breast cancer cells with enhanced endoplasmic reticulum stress. PLoS One 12: e0176348 , 2017. doi:10.1371/journal.pone.0176348.
    Crossref | PubMed | ISI | Google Scholar
  • 167. Lum JJ, Bauer DE, Kong M, Harris MH, Li C, Lindsten T, Thompson CB. Growth factor regulation of autophagy and cell survival in the absence of apoptosis. Cell 120: 237–248, 2005. doi:10.1016/j.cell.2004.11.046.
    Crossref | PubMed | ISI | Google Scholar
  • 168. Luo S, Rubinsztein DC. Apoptosis blocks Beclin 1-dependent autophagosome synthesis: an effect rescued by Bcl-xL. Cell Death Differ 17: 268–277, 2010. doi:10.1038/cdd.2009.121.
    Crossref | PubMed | ISI | Google Scholar
  • 169. Lüthi AU, Martin SJ. The CASBAH: a searchable database of caspase substrates. Cell Death Differ 14: 641–650, 2007. doi:10.1038/sj.cdd.4402103.
    Crossref | PubMed | ISI | Google Scholar
  • 170. MacCormac LP, Muqit MM, Faulkes DJ, Wood NW, Latchman DS. Reduction in endogenous parkin levels renders glial cells sensitive to both caspase-dependent and caspase-independent cell death. Eur J Neurosci 20: 2038–2048, 2004. doi:10.1111/j.1460-9568.2004.03659.x.
    Crossref | PubMed | ISI | Google Scholar
  • 171. Madeo F, Zimmermann A, Maiuri MC, Kroemer G. Essential role for autophagy in life span extension. J Clin Invest 125: 85–93, 2015. doi:10.1172/JCI73946.
    Crossref | PubMed | ISI | Google Scholar
  • 172. Maiuri MC, Le Toumelin G, Criollo A, Rain JC, Gautier F, Juin P, Tasdemir E, Pierron G, Troulinaki K, Tavernarakis N, Hickman JA, Geneste O, Kroemer G. Functional and physical interaction between Bcl-X(L) and a BH3-like domain in Beclin-1. EMBO J 26: 2527–2539, 2007. doi:10.1038/sj.emboj.7601689.
    Crossref | PubMed | ISI | Google Scholar
  • 173. Mammucari C, Milan G, Romanello V, Masiero E, Rudolf R, Del Piccolo P, Burden SJ, Di Lisi R, Sandri C, Zhao J, Goldberg AL, Schiaffino S, Sandri M. FoxO3 controls autophagy in skeletal muscle in vivo. Cell Metab 6: 458–471, 2007. doi:10.1016/j.cmet.2007.11.001.
    Crossref | PubMed | ISI | Google Scholar
  • 174. Mariño G, Niso-Santano M, Baehrecke EH, Kroemer G. Self-consumption: the interplay of autophagy and apoptosis. Nat Rev Mol Cell Biol 15: 81–94, 2014. doi:10.1038/nrm3735.
    Crossref | PubMed | ISI | Google Scholar
  • 175. Masiero E, Agatea L, Mammucari C, Blaauw B, Loro E, Komatsu M, Metzger D, Reggiani C, Schiaffino S, Sandri M. Autophagy is required to maintain muscle mass. Cell Metab 10: 507–515, 2009. doi:10.1016/j.cmet.2009.10.008.
    Crossref | PubMed | ISI | Google Scholar
  • 176. Mathew R, Karp CM, Beaudoin B, Vuong N, Chen G, Chen HY, Bray K, Reddy A, Bhanot G, Gelinas C, Dipaola RS, Karantza-Wadsworth V, White E. Autophagy suppresses tumorigenesis through elimination of p62. Cell 137: 1062–1075, 2009. doi:10.1016/j.cell.2009.03.048.
    Crossref | PubMed | ISI | Google Scholar
  • 177. Mathew R, Kongara S, Beaudoin B, Karp CM, Bray K, Degenhardt K, Chen G, Jin S, White E. Autophagy suppresses tumor progression by limiting chromosomal instability. Genes Dev 21: 1367–1381, 2007. doi:10.1101/gad.1545107.
    Crossref | PubMed | ISI | Google Scholar
  • 178. Mattison JA, Colman RJ, Beasley TM, Allison DB, Kemnitz JW, Roth GS, Ingram DK, Weindruch R, de Cabo R, Anderson RM. Caloric restriction improves health and survival of rhesus monkeys. Nat Commun 8: 14063, 2017. doi:10.1038/ncomms14063.
    Crossref | PubMed | ISI | Google Scholar
  • 179. McLelland GL, Soubannier V, Chen CX, McBride HM, Fon EA. Parkin and PINK1 function in a vesicular trafficking pathway regulating mitochondrial quality control. EMBO J 33: 282–295, 2014. doi:10.1002/embj.201385902.
    Crossref | PubMed | ISI | Google Scholar
  • 180. McMillan EM, Quadrilatero J. Autophagy is required and protects against apoptosis during myoblast differentiation. Biochem J 462: 267–277, 2014. doi:10.1042/BJ20140312.
    Crossref | PubMed | ISI | Google Scholar
  • 181. Mewton N, Croisille P, Gahide G, Rioufol G, Bonnefoy E, Sanchez I, Cung TT, Sportouch C, Angoulvant D, Finet G, André-Fouët X, Derumeaux G, Piot C, Vernhet H, Revel D, Ovize M. Effect of cyclosporine on left ventricular remodeling after reperfused myocardial infarction. J Am Coll Cardiol 55: 1200–1205, 2010. doi:10.1016/j.jacc.2009.10.052.
    Crossref | PubMed | ISI | Google Scholar
  • 182. Michiorri S, Gelmetti V, Giarda E, Lombardi F, Romano F, Marongiu R, Nerini-Molteni S, Sale P, Vago R, Arena G, Torosantucci L, Cassina L, Russo MA, Dallapiccola B, Valente EM, Casari G. The Parkinson-associated protein PINK1 interacts with Beclin1 and promotes autophagy. Cell Death Differ 17: 962–974, 2010. doi:10.1038/cdd.2009.200.
    Crossref | PubMed | ISI | Google Scholar
  • 183. Mizushima N, Kuma A, Kobayashi Y, Yamamoto A, Matsubae M, Takao T, Natsume T, Ohsumi Y, Yoshimori T. Mouse Apg16L, a novel WD-repeat protein, targets to the autophagic isolation membrane with the Apg12-Apg5 conjugate. J Cell Sci 116: 1679–1688, 2003. doi:10.1242/jcs.00381.
    Crossref | PubMed | ISI | Google Scholar
  • 184. Mizushima N, Yamamoto A, Matsui M, Yoshimori T, Ohsumi Y. In vivo analysis of autophagy in response to nutrient starvation using transgenic mice expressing a fluorescent autophagosome marker. Mol Biol Cell 15: 1101–1111, 2004. doi:10.1091/mbc.e03-09-0704.
    Crossref | PubMed | ISI | Google Scholar
  • 185. Møller AB, Kampmann U, Hedegaard J, Thorsen K, Nordentoft I, Vendelbo MH, Møller N, Jessen N. Altered gene expression and repressed markers of autophagy in skeletal muscle of insulin resistant patients with type 2 diabetes. Sci Rep 7: 43775, 2017. doi:10.1038/srep43775.
    Crossref | PubMed | ISI | Google Scholar
  • 186. Mordier S, Deval C, Béchet D, Tassa A, Ferrara M. Leucine limitation induces autophagy and activation of lysosome-dependent proteolysis in C2C12 myotubes through a mammalian target of rapamycin-independent signaling pathway. J Biol Chem 275: 29900–29906, 2000. doi:10.1074/jbc.M003633200.
    Crossref | PubMed | ISI | Google Scholar
  • 187. Morselli E, Shen S, Ruckenstuhl C, Bauer MA, Mariño G, Galluzzi L, Criollo A, Michaud M, Maiuri MC, Chano T, Madeo F, Kroemer G. p53 inhibits autophagy by interacting with the human ortholog of yeast Atg17, RB1CC1/FIP200. Cell Cycle 10: 2763–2769, 2011. doi:10.4161/cc.10.16.16868.
    Crossref | PubMed | ISI | Google Scholar
  • 188. Murach KA, Englund DA, Dupont-Versteegden EE, McCarthy JJ, Peterson CA. Myonuclear domain flexibility challenges rigid assumptions on satellite cell contribution to skeletal muscle fiber hypertrophy. Front Physiol 9: 635, 2018. doi:10.3389/fphys.2018.00635.
    Crossref | PubMed | ISI | Google Scholar
  • 189. Murakawa T, Yamaguchi O, Hashimoto A, Hikoso S, Takeda T, Oka T, Yasui H, Ueda H, Akazawa Y, Nakayama H, Taneike M, Misaka T, Omiya S, Shah AM, Yamamoto A, Nishida K, Ohsumi Y, Okamoto K, Sakata Y, Otsu K. Bcl-2-like protein 13 is a mammalian Atg32 homologue that mediates mitophagy and mitochondrial fragmentation. Nat Commun 6: 7527, 2015. doi:10.1038/ncomms8527.
    Crossref | PubMed | ISI | Google Scholar
  • 190. Namba T, Takabatake Y, Kimura T, Takahashi A, Yamamoto T, Matsuda J, Kitamura H, Niimura F, Matsusaka T, Iwatani H, Matsui I, Kaimori J, Kioka H, Isaka Y, Rakugi H. Autophagic clearance of mitochondria in the kidney copes with metabolic acidosis. J Am Soc Nephrol 25: 2254–2266, 2014. doi:10.1681/ASN.2013090986.
    Crossref | PubMed | ISI | Google Scholar
  • 191. Narendra D, Kane LA, Hauser DN, Fearnley IM, Youle RJ. p62/SQSTM1 is required for Parkin-induced mitochondrial clustering but not mitophagy; VDAC1 is dispensable for both. Autophagy 6: 1090–1106, 2010. doi:10.4161/auto.6.8.13426.
    Crossref | PubMed | ISI | Google Scholar
  • 192. Narendra D, Tanaka A, Suen DF, Youle RJ. Parkin is recruited selectively to impaired mitochondria and promotes their autophagy. J Cell Biol 183: 795–803, 2008. doi:10.1083/jcb.200809125.
    Crossref | PubMed | ISI | Google Scholar
  • 193. Narendra DP, Jin SM, Tanaka A, Suen DF, Gautier CA, Shen J, Cookson MR, Youle RJ. PINK1 is selectively stabilized on impaired mitochondria to activate Parkin. PLoS Biol 8: e1000298 , 2010. doi:10.1371/journal.pbio.1000298.
    Crossref | PubMed | ISI | Google Scholar
  • 194. Nishida Y, Arakawa S, Fujitani K, Yamaguchi H, Mizuta T, Kanaseki T, Komatsu M, Otsu K, Tsujimoto Y, Shimizu S. Discovery of Atg5/Atg7-independent alternative macroautophagy. Nature 461: 654–658, 2009. [Erratum in Nature 533: 130, 2016.] doi:10.1038/nature08455.
    Crossref | PubMed | ISI | Google Scholar
  • 195. Novak I, Kirkin V, McEwan DG, Zhang J, Wild P, Rozenknop A, Rogov V, Löhr F, Popovic D, Occhipinti A, Reichert AS, Terzic J, Dötsch V, Ney PA, Dikic I. Nix is a selective autophagy receptor for mitochondrial clearance. EMBO Rep 11: 45–51, 2010. doi:10.1038/embor.2009.256.
    Crossref | PubMed | ISI | Google Scholar
  • 196. Ogata T, Oishi Y, Higuchi M, Muraoka I. Fasting-related autophagic response in slow- and fast-twitch skeletal muscle. Biochem Biophys Res Commun 394: 136–140, 2010. doi:10.1016/j.bbrc.2010.02.130.
    Crossref | PubMed | ISI | Google Scholar
  • 197. Okatsu K, Saisho K, Shimanuki M, Nakada K, Shitara H, Sou YS, Kimura M, Sato S, Hattori N, Komatsu M, Tanaka K, Matsuda N. p62/SQSTM1 cooperates with Parkin for perinuclear clustering of depolarized mitochondria. Genes Cells 15: 887–900, 2010. doi:10.1111/j.1365-2443.2010.01426.x.
    Crossref | PubMed | ISI | Google Scholar
  • 198. O’Leary MF, Vainshtein A, Carter HN, Zhang Y, Hood DA. Denervation-induced mitochondrial dysfunction and autophagy in skeletal muscle of apoptosis-deficient animals. Am J Physiol Cell Physiol 303: C447–C454, 2012. doi:10.1152/ajpcell.00451.2011.
    Link | ISI | Google Scholar
  • 199. O’Leary MF, Vainshtein A, Iqbal S, Ostojic O, Hood DA. Adaptive plasticity of autophagic proteins to denervation in aging skeletal muscle. Am J Physiol Cell Physiol 304: C422–C430, 2013. doi:10.1152/ajpcell.00240.2012.
    Link | ISI | Google Scholar
  • 200. O’Neill KL, Huang K, Zhang J, Chen Y, Luo X. Inactivation of prosurvival Bcl-2 proteins activates Bax/Bak through the outer mitochondrial membrane. Genes Dev 30: 973–988, 2016. doi:10.1101/gad.276725.115.
    Crossref | PubMed | ISI | Google Scholar
  • 201. Oral O, Oz-Arslan D, Itah Z, Naghavi A, Deveci R, Karacali S, Gozuacik D. Cleavage of Atg3 protein by caspase-8 regulates autophagy during receptor-activated cell death. Apoptosis 17: 810–820, 2012. doi:10.1007/s10495-012-0735-0.
    Crossref | PubMed | ISI | Google Scholar
  • 202. Ott M, Robertson JD, Gogvadze V, Zhivotovsky B, Orrenius S. Cytochrome c release from mitochondria proceeds by a two-step process. Proc Natl Acad Sci USA 99: 1259–1263, 2002. doi:10.1073/pnas.241655498.
    Crossref | PubMed | ISI | Google Scholar
  • 203. Pagliarini V, Wirawan E, Romagnoli A, Ciccosanti F, Lisi G, Lippens S, Cecconi F, Fimia GM, Vandenabeele P, Corazzari M, Piacentini M. Proteolysis of Ambra1 during apoptosis has a role in the inhibition of the autophagic pro-survival response. Cell Death Differ 19: 1495–1504, 2012. doi:10.1038/cdd.2012.27.
    Crossref | PubMed | ISI | Google Scholar
  • 204. Palikaras K, Lionaki E, Tavernarakis N. Mechanisms of mitophagy in cellular homeostasis, physiology and pathology. Nat Cell Biol 20: 1013–1022, 2018. doi:10.1038/s41556-018-0176-2.
    Crossref | PubMed | ISI | Google Scholar
  • 205. Pankiv S, Clausen TH, Lamark T, Brech A, Bruun JA, Outzen H, Øvervatn A, Bjørkøy G, Johansen T. p62/SQSTM1 binds directly to Atg8/LC3 to facilitate degradation of ubiquitinated protein aggregates by autophagy. J Biol Chem 282: 24131–24145, 2007. doi:10.1074/jbc.M702824200.
    Crossref | PubMed | ISI | Google Scholar
  • 206. Paré MF, Baechler BL, Fajardo VA, Earl E, Wong E, Campbell TL, Tupling AR, Quadrilatero J. Effect of acute and chronic autophagy deficiency on skeletal muscle apoptotic signaling, morphology, and function. Biochim Biophys Acta Mol Cell Res 1864: 708–718, 2017. doi:10.1016/j.bbamcr.2016.12.015.
    Crossref | PubMed | ISI | Google Scholar
  • 207. Pattingre S, Tassa A, Qu X, Garuti R, Liang XH, Mizushima N, Packer M, Schneider MD, Levine B. Bcl-2 antiapoptotic proteins inhibit Beclin 1-dependent autophagy. Cell 122: 927–939, 2005. doi:10.1016/j.cell.2005.07.002.
    Crossref | PubMed | ISI | Google Scholar
  • 208. Piétri-Rouxel F, Gentil C, Vassilopoulos S, Baas D, Mouisel E, Ferry A, Vignaud A, Hourdé C, Marty I, Schaeffer L, Voit T, Garcia L. DHPR alpha1S subunit controls skeletal muscle mass and morphogenesis. EMBO J 29: 643–654, 2010. doi:10.1038/emboj.2009.366.
    Crossref | PubMed | ISI | Google Scholar
  • 209. Pigna E, Sanna K, Coletti D, Li Z, Parlakian A, Adamo S, Moresi V. Increasing autophagy does not affect neurogenic muscle atrophy. Eur J Transl Myol 28: 7687, 2018. doi:10.4081/ejtm.2018.7687.
    Crossref | PubMed | ISI | Google Scholar
  • 210. Piot C, Croisille P, Staat P, Thibault H, Rioufol G, Mewton N, Elbelghiti R, Cung TT, Bonnefoy E, Angoulvant D, Macia C, Raczka F, Sportouch C, Gahide G, Finet G, André-Fouët X, Revel D, Kirkorian G, Monassier JP, Derumeaux G, Ovize M. Effect of cyclosporine on reperfusion injury in acute myocardial infarction. N Engl J Med 359: 473–481, 2008. doi:10.1056/NEJMoa071142.
    Crossref | PubMed | ISI | Google Scholar
  • 211. Plant PJ, Bain JR, Correa JE, Woo M, Batt J. Absence of caspase-3 protects against denervation-induced skeletal muscle atrophy. J Appl Physiol (1985) 107: 224–234, 2009. doi:10.1152/japplphysiol.90932.2008.
    Link | ISI | Google Scholar
  • 212. Polson HE, de Lartigue J, Rigden DJ, Reedijk M, Urbé S, Clague MJ, Tooze SA. Mammalian Atg18 (WIPI2) localizes to omegasome-anchored phagophores and positively regulates LC3 lipidation. Autophagy 6: 506–522, 2010. doi:10.4161/auto.6.4.11863.
    Crossref | PubMed | ISI | Google Scholar
  • 213. Quadrilatero J, Alway SE, Dupont-Versteegden EE. Skeletal muscle apoptotic response to physical activity: potential mechanisms for protection. Appl Physiol Nutr Metab 36: 608–617, 2011. doi:10.1139/h11-064.
    Crossref | PubMed | ISI | Google Scholar
  • 214. Ravikumar B, Moreau K, Jahreiss L, Puri C, Rubinsztein DC. Plasma membrane contributes to the formation of pre-autophagosomal structures. Nat Cell Biol 12: 747–757, 2010. [Erratum in Nat Cell Biol 12: 1021, 2010.] doi:10.1038/ncb2078.
    Crossref | PubMed | ISI | Google Scholar
  • 215. Ravikumar B, Sarkar S, Davies JE, Futter M, Garcia-Arencibia M, Green-Thompson ZW, Jimenez-Sanchez M, Korolchuk VI, Lichtenberg M, Luo S, Massey DC, Menzies FM, Moreau K, Narayanan U, Renna M, Siddiqi FH, Underwood BR, Winslow AR, Rubinsztein DC. Regulation of mammalian autophagy in physiology and pathophysiology. Physiol Rev 90: 1383–1435, 2010. doi:10.1152/physrev.00030.2009.
    Link | ISI | Google Scholar
  • 216. Ren Y, Li Y, Yan J, Ma M, Zhou D, Xue Z, Zhang Z, Liu H, Yang H, Jia L, Zhang L, Zhang Q, Mu S, Zhang R, Da Y. Adiponectin modulates oxidative stress-induced mitophagy and protects C2C12 myoblasts against apoptosis. Sci Rep 7: 3209, 2017. doi:10.1038/s41598-017-03319-2.
    Crossref | PubMed | ISI | Google Scholar
  • 217. Richter B, Sliter DA, Herhaus L, Stolz A, Wang C, Beli P, Zaffagnini G, Wild P, Martens S, Wagner SA, Youle RJ, Dikic I. Phosphorylation of OPTN by TBK1 enhances its binding to Ub chains and promotes selective autophagy of damaged mitochondria. Proc Natl Acad Sci USA 113: 4039–4044, 2016. doi:10.1073/pnas.1523926113.
    Crossref | PubMed | ISI | Google Scholar
  • 218. Rikka S, Quinsay MN, Thomas RL, Kubli DA, Zhang X, Murphy AN, Gustafsson AB. Bnip3 impairs mitochondrial bioenergetics and stimulates mitochondrial turnover. Cell Death Differ 18: 721–731, 2011. doi:10.1038/cdd.2010.146.
    Crossref | PubMed | ISI | Google Scholar
  • 219. Roberts RF, Tang MY, Fon EA, Durcan TM. Defending the mitochondria: the pathways of mitophagy and mitochondrial-derived vesicles. Int J Biochem Cell Biol 79: 427–436, 2016. doi:10.1016/j.biocel.2016.07.020.
    Crossref | PubMed | ISI | Google Scholar
  • 220. Rosen KM, Moussa CE, Lee HK, Kumar P, Kitada T, Qin G, Fu Q, Querfurth HW. Parkin reverses intracellular beta-amyloid accumulation and its negative effects on proteasome function. J Neurosci Res 88: 167–178, 2010. doi:10.1002/jnr.22178.
    Crossref | PubMed | ISI | Google Scholar
  • 221. Rubinstein AD, Eisenstein M, Ber Y, Bialik S, Kimchi A. The autophagy protein Atg12 associates with antiapoptotic Bcl-2 family members to promote mitochondrial apoptosis. Mol Cell 44: 698–709, 2011. doi:10.1016/j.molcel.2011.10.014.
    Crossref | PubMed | ISI | Google Scholar
  • 222. Ryu D, Mouchiroud L, Andreux PA, Katsyuba E, Moullan N, Nicolet-Dit-Félix AA, Williams EG, Jha P, Lo Sasso G, Huzard D, Aebischer P, Sandi C, Rinsch C, Auwerx J. Urolithin A induces mitophagy and prolongs lifespan in C. elegans and increases muscle function in rodents. Nat Med 22: 879–888, 2016. doi:10.1038/nm.4132.
    Crossref | PubMed | ISI | Google Scholar
  • 223. Sachewsky N, Hunt J, Cooke MJ, Azimi A, Zarin T, Miu C, Shoichet MS, Morshead CM. Cyclosporin A enhances neural precursor cell survival in mice through a calcineurin-independent pathway. Dis Model Mech 7: 953–961, 2014. doi:10.1242/dmm.014480.
    Crossref | PubMed | ISI | Google Scholar
  • 224. Saikumar P, Dong Z, Mikhailov V, Denton M, Weinberg JM, Venkatachalam MA. Apoptosis: definition, mechanisms, and relevance to disease. Am J Med 107: 489–506, 1999. doi:10.1016/S0002-9343(99)00259-4.
    Crossref | PubMed | ISI | Google Scholar
  • 225. Saitoh T, Fujita N, Hayashi T, Takahara K, Satoh T, Lee H, Matsunaga K, Kageyama S, Omori H, Noda T, Yamamoto N, Kawai T, Ishii K, Takeuchi O, Yoshimori T, Akira S. Atg9a controls dsDNA-driven dynamic translocation of STING and the innate immune response. Proc Natl Acad Sci USA 106: 20842–20846, 2009. doi:10.1073/pnas.0911267106.
    Crossref | PubMed | ISI | Google Scholar
  • 226. Saitoh T, Fujita N, Jang MH, Uematsu S, Yang BG, Satoh T, Omori H, Noda T, Yamamoto N, Komatsu M, Tanaka K, Kawai T, Tsujimura T, Takeuchi O, Yoshimori T, Akira S. Loss of the autophagy protein Atg16L1 enhances endotoxin-induced IL-1beta production. Nature 456: 264–268, 2008. doi:10.1038/nature07383.
    Crossref | PubMed | ISI | Google Scholar
  • 227. Sakuma K, Kinoshita M, Ito Y, Aizawa M, Aoi W, Yamaguchi A. p62/SQSTM1 but not LC3 is accumulated in sarcopenic muscle of mice. J Cachexia Sarcopenia Muscle 7: 204–212, 2016. doi:10.1002/jcsm.12045.
    Crossref | PubMed | ISI | Google Scholar
  • 228. Salminen A, Vihko V. Autophagic response to strenuous exercise in mouse skeletal muscle fibers. Virchows Arch B Cell Pathol Incl Mol Pathol 45: 97–106, 1984. doi:10.1007/BF02889856.
    Crossref | PubMed | Google Scholar
  • 229. Sancak Y, Bar-Peled L, Zoncu R, Markhard AL, Nada S, Sabatini DM. Ragulator-Rag complex targets mTORC1 to the lysosomal surface and is necessary for its activation by amino acids. Cell 141: 290–303, 2010. doi:10.1016/j.cell.2010.02.024.
    Crossref | PubMed | ISI | Google Scholar
  • 230. Sancak Y, Peterson TR, Shaul YD, Lindquist RA, Thoreen CC, Bar-Peled L, Sabatini DM. The Rag GTPases bind raptor and mediate amino acid signaling to mTORC1. Science 320: 1496–1501, 2008. doi:10.1126/science.1157535.
    Crossref | PubMed | ISI | Google Scholar
  • 231. Sandoval H, Thiagarajan P, Dasgupta SK, Schumacher A, Prchal JT, Chen M, Wang J. Essential role for Nix in autophagic maturation of erythroid cells. Nature 454: 232–235, 2008. doi:10.1038/nature07006.
    Crossref | PubMed | ISI | Google Scholar
  • 232. Sandri M. Autophagy in skeletal muscle. FEBS Lett 584: 1411–1416, 2010. doi:10.1016/j.febslet.2010.01.056.
    Crossref | PubMed | ISI | Google Scholar
  • 233. Sandri M, El Meslemani AH, Sandri C, Schjerling P, Vissing K, Andersen JL, Rossini K, Carraro U, Angelini C. Caspase 3 expression correlates with skeletal muscle apoptosis in Duchenne and facioscapulo human muscular dystrophy. A potential target for pharmacological treatment? J Neuropathol Exp Neurol 60: 302–312, 2001. doi:10.1093/jnen/60.3.302.
    Crossref | PubMed | ISI | Google Scholar
  • 234. Schakman O, Dehoux M, Bouchuari S, Delaere S, Lause P, Decroly N, Shoelson SE, Thissen JP. Role of IGF-I and the TNFα/NF-κB pathway in the induction of muscle atrogenes by acute inflammation. Am J Physiol Endocrinol Metab 303: E729–E739, 2012. doi:10.1152/ajpendo.00060.2012.
    Link | ISI | Google Scholar
  • 235. Scherz-Shouval R, Elazar Z. Regulation of autophagy by ROS: physiology and pathology. Trends Biochem Sci 36: 30–38, 2011. doi:10.1016/j.tibs.2010.07.007.
    Crossref | PubMed | ISI | Google Scholar
  • 236. Scherz-Shouval R, Shvets E, Fass E, Shorer H, Gil L, Elazar Z. Reactive oxygen species are essential for autophagy and specifically regulate the activity of Atg4. EMBO J 26: 1749–1760, 2007. doi:10.1038/sj.emboj.7601623.
    Crossref | PubMed | ISI | Google Scholar
  • 237. Schieber M, Chandel NS. ROS function in redox signaling and oxidative stress. Curr Biol 24: R453–R462, 2014. doi:10.1016/j.cub.2014.03.034.
    Crossref | PubMed | ISI | Google Scholar
  • 238. Schwarten M, Mohrlüder J, Ma P, Stoldt M, Thielmann Y, Stangler T, Hersch N, Hoffmann B, Merkel R, Willbold D. Nix directly binds to GABARAP: a possible crosstalk between apoptosis and autophagy. Autophagy 5: 690–698, 2009. doi:10.4161/auto.5.5.8494.
    Crossref | PubMed | ISI | Google Scholar
  • 239. Schwartz LM. Skeletal muscles do not undergo apoptosis during either atrophy or programmed cell death-revisiting the myonuclear domain hypothesis. Front Physiol 9: 1887, 2019. doi:10.3389/fphys.2018.01887.
    Crossref | PubMed | ISI | Google Scholar
  • 240. Shiba-Fukushima K, Arano T, Matsumoto G, Inoshita T, Yoshida S, Ishihama Y, Ryu KY, Nukina N, Hattori N, Imai Y. Phosphorylation of mitochondrial polyubiquitin by PINK1 promotes Parkin mitochondrial tethering. PLoS Genet 10: e1004861 , 2014. doi:10.1371/journal.pgen.1004861.
    Crossref | PubMed | ISI | Google Scholar
  • 241. Simm A, Bertsch G, Frank H, Zimmermann U, Hoppe J. Cell death of AKR-2B fibroblasts after serum removal: a process between apoptosis and necrosis. J Cell Sci 110: 819–828, 1997.
    Crossref | PubMed | ISI | Google Scholar
  • 242. Sin J, Andres AM, Taylor DJ, Weston T, Hiraumi Y, Stotland A, Kim BJ, Huang C, Doran KS, Gottlieb RA. Mitophagy is required for mitochondrial biogenesis and myogenic differentiation of C2C12 myoblasts. Autophagy 12: 369–380, 2016. doi:10.1080/15548627.2015.1115172.
    Crossref | PubMed | ISI | Google Scholar
  • 243. Sinha K, Das J, Pal PB, Sil PC. Oxidative stress: the mitochondria-dependent and mitochondria-independent pathways of apoptosis. Arch Toxicol 87: 1157–1180, 2013. doi:10.1007/s00204-013-1034-4.
    Crossref | PubMed | ISI | Google Scholar
  • 244. Siu PM, Alway SE. Deficiency of the Bax gene attenuates denervation-induced apoptosis. Apoptosis 11: 967–981, 2006. doi:10.1007/s10495-006-6315-4.
    Crossref | PubMed | ISI | Google Scholar
  • 245. Song M, Gong G, Burelle Y, Gustafsson AB, Kitsis RN, Matkovich SJ, Dorn GW II. Interdependence of Parkin-mediated mitophagy and mitochondrial fission in adult mouse hearts. Circ Res 117: 346–351, 2015. doi:10.1161/CIRCRESAHA.117.306859.
    Crossref | PubMed | ISI | Google Scholar
  • 246. Sou YS, Waguri S, Iwata J, Ueno T, Fujimura T, Hara T, Sawada N, Yamada A, Mizushima N, Uchiyama Y, Kominami E, Tanaka K, Komatsu M. The Atg8 conjugation system is indispensable for proper development of autophagic isolation membranes in mice. Mol Biol Cell 19: 4762–4775, 2008. doi:10.1091/mbc.e08-03-0309.
    Crossref | PubMed | ISI | Google Scholar
  • 247. Soubannier V, McLelland GL, Zunino R, Braschi E, Rippstein P, Fon EA, McBride HM. A vesicular transport pathway shuttles cargo from mitochondria to lysosomes. Curr Biol 22: 135–141, 2012. doi:10.1016/j.cub.2011.11.057.
    Crossref | PubMed | ISI | Google Scholar
  • 248. Stolz A, Ernst A, Dikic I. Cargo recognition and trafficking in selective autophagy. Nat Cell Biol 16: 495–501, 2014. doi:10.1038/ncb2979.
    Crossref | PubMed | ISI | Google Scholar
  • 249. Strappazzon F, Nazio F, Corrado M, Cianfanelli V, Romagnoli A, Fimia GM, Campello S, Nardacci R, Piacentini M, Campanella M, Cecconi F. AMBRA1 is able to induce mitophagy via LC3 binding, regardless of PARKIN and p62/SQSTM1. Cell Death Differ 22: 419–432, 2015. [Erratum in Cell Death Differ 22: 517, 2015. 10.1038/cdd.2014.190. 25661525] doi:10.1038/cdd.2014.139.
    Crossref | PubMed | ISI | Google Scholar
  • 250. Sun Y, Vashisht AA, Tchieu J, Wohlschlegel JA, Dreier L. Voltage-dependent anion channels (VDACs) recruit Parkin to defective mitochondria to promote mitochondrial autophagy. J Biol Chem 287: 40652–40660, 2012. doi:10.1074/jbc.M112.419721.
    Crossref | PubMed | ISI | Google Scholar
  • 251. Susin SA, Zamzami N, Castedo M, Hirsch T, Marchetti P, Macho A, Daugas E, Geuskens M, Kroemer G. Bcl-2 inhibits the mitochondrial release of an apoptogenic protease. J Exp Med 184: 1331–1341, 1996. doi:10.1084/jem.184.4.1331.
    Crossref | PubMed | ISI | Google Scholar
  • 252. Taillandier D, Aurousseau E, Meynial-Denis D, Bechet D, Ferrara M, Cottin P, Ducastaing A, Bigard X, Guezennec CY, Schmid HP, Attaix D. Coordinate activation of lysosomal, Ca 2+-activated and ATP-ubiquitin-dependent proteinases in the unweighted rat soleus muscle. Biochem J 316: 65–72, 1996. doi:10.1042/bj3160065.
    Crossref | PubMed | ISI | Google Scholar
  • 253. Tanaka A, Cleland MM, Xu S, Narendra DP, Suen DF, Karbowski M, Youle RJ. Proteasome and p97 mediate mitophagy and degradation of mitofusins induced by Parkin. J Cell Biol 191: 1367–1380, 2010. doi:10.1083/jcb.201007013.
    Crossref | PubMed | ISI | Google Scholar
  • 254. Tanaka Y, Guhde G, Suter A, Eskelinen EL, Hartmann D, Lüllmann-Rauch R, Janssen PM, Blanz J, von Figura K, Saftig P. Accumulation of autophagic vacuoles and cardiomyopathy in LAMP-2-deficient mice. Nature 406: 902–906, 2000. doi:10.1038/35022595.
    Crossref | PubMed | ISI | Google Scholar
  • 255. Tanida I, Sou YS, Ezaki J, Minematsu-Ikeguchi N, Ueno T, Kominami E. HsAtg4B/HsApg4B/autophagin-1 cleaves the carboxyl termini of three human Atg8 homologues and delipidates microtubule-associated protein light chain 3- and GABAA receptor-associated protein-phospholipid conjugates. J Biol Chem 279: 36268–36276, 2004. doi:10.1074/jbc.M401461200.
    Crossref | PubMed | ISI | Google Scholar
  • 256. Tasdemir E, Maiuri MC, Galluzzi L, Vitale I, Djavaheri-Mergny M, D’Amelio M, Criollo A, Morselli E, Zhu C, Harper F, Nannmark U, Samara C, Pinton P, Vicencio JM, Carnuccio R, Moll UM, Madeo F, Paterlini-Brechot P, Rizzuto R, Szabadkai G, Pierron G, Blomgren K, Tavernarakis N, Codogno P, Cecconi F, Kroemer G. Regulation of autophagy by cytoplasmic p53. Nat Cell Biol 10: 676–687, 2008. doi:10.1038/ncb1730.
    Crossref | PubMed | ISI | Google Scholar
  • 257. Tassa A, Roux MP, Attaix D, Bechet DM. Class III phosphoinositide 3-kinase--Beclin1 complex mediates the amino acid-dependent regulation of autophagy in C2C12 myotubes. Biochem J 376: 577–586, 2003. doi:10.1042/bj20030826.
    Crossref | PubMed | ISI | Google Scholar
  • 258. Tidball JG, Albrecht DE, Lokensgard BE, Spencer MJ. Apoptosis precedes necrosis of dystrophin-deficient muscle. J Cell Sci 108: 2197–2204, 1995.
    Crossref | PubMed | ISI | Google Scholar
  • 259. Tracy K, Dibling BC, Spike BT, Knabb JR, Schumacker P, Macleod KF. BNIP3 is an RB/E2F target gene required for hypoxia-induced autophagy. Mol Cell Biol 27: 6229–6242, 2007. doi:10.1128/MCB.02246-06.
    Crossref | PubMed | ISI | Google Scholar
  • 260. Troncoso R, Paredes F, Parra V, Gatica D, Vásquez-Trincado C, Quiroga C, Bravo-Sagua R, López-Crisosto C, Rodriguez AE, Oyarzún AP, Kroemer G, Lavandero S. Dexamethasone-induced autophagy mediates muscle atrophy through mitochondrial clearance. Cell Cycle 13: 2281–2295, 2014. doi:10.4161/cc.29272.
    Crossref | PubMed | ISI | Google Scholar
  • 261. Twig G, Elorza A, Molina AJ, Mohamed H, Wikstrom JD, Walzer G, Stiles L, Haigh SE, Katz S, Las G, Alroy J, Wu M, Py BF, Yuan J, Deeney JT, Corkey BE, Shirihai OS. Fission and selective fusion govern mitochondrial segregation and elimination by autophagy. EMBO J 27: 433–446, 2008. doi:10.1038/sj.emboj.7601963.
    Crossref | PubMed | ISI | Google Scholar
  • 262. Urfer SR, Kaeberlein TL, Mailheau S, Bergman PJ, Creevy KE, Promislow DEL, Kaeberlein M. A randomized controlled trial to establish effects of short-term rapamycin treatment in 24 middle-aged companion dogs. Geroscience 39: 117–127, 2017. doi:10.1007/s11357-017-9972-z.
    Crossref | PubMed | ISI | Google Scholar
  • 263. Valente EM, Abou-Sleiman PM, Caputo V, Muqit MM, Harvey K, Gispert S, Ali Z, Del Turco D, Bentivoglio AR, Healy DG, Albanese A, Nussbaum R, González-Maldonado R, Deller T, Salvi S, Cortelli P, Gilks WP, Latchman DS, Harvey RJ, Dallapiccola B, Auburger G, Wood NW. Hereditary early-onset Parkinson’s disease caused by mutations in PINK1. Science 304: 1158–1160, 2004. doi:10.1126/science.1096284.
    Crossref | PubMed | ISI | Google Scholar
  • 264. Van Humbeeck C, Cornelissen T, Hofkens H, Mandemakers W, Gevaert K, De Strooper B, Vandenberghe W. Parkin interacts with Ambra1 to induce mitophagy. J Neurosci 31: 10249–10261, 2011. doi:10.1523/JNEUROSCI.1917-11.2011.
    Crossref | PubMed | ISI | Google Scholar
  • 265. Vandenabeele P, Galluzzi L, Vanden Berghe T, Kroemer G. Molecular mechanisms of necroptosis: an ordered cellular explosion. Nat Rev Mol Cell Biol 11: 700–714, 2010. doi:10.1038/nrm2970.
    Crossref | PubMed | ISI | Google Scholar
  • 266. Vanhorebeek I, Gunst J, Derde S, Derese I, Boussemaere M, Güiza F, Martinet W, Timmermans JP, D’Hoore A, Wouters PJ, Van den Berghe G. Insufficient activation of autophagy allows cellular damage to accumulate in critically ill patients. J Clin Endocrinol Metab 96: E633–E645, 2011. doi:10.1210/jc.2010-2563.
    Crossref | PubMed | ISI | Google Scholar
  • 267. Vinel C, Lukjanenko L, Batut A, Deleruyelle S, Pradère JP, Le Gonidec S, Dortignac A, Geoffre N, Pereira O, Karaz S, Lee U, Camus M, Chaoui K, Mouisel E, Bigot A, Mouly V, Vigneau M, Pagano AF, Chopard A, Pillard F, Guyonnet S, Cesari M, Burlet-Schiltz O, Pahor M, Feige JN, Vellas B, Valet P, Dray C. The exerkine apelin reverses age-associated sarcopenia. Nat Med 24: 1360–1371, 2018. doi:10.1038/s41591-018-0131-6.
    Crossref | PubMed | ISI | Google Scholar
  • 268. Wang B, Lu D, Xuan M, Hu W. Antitumor effect of sunitinib in human prostate cancer cells functions via autophagy. Exp Ther Med 13: 1285–1294, 2017. doi:10.3892/etm.2017.4134.
    Crossref | PubMed | ISI | Google Scholar
  • 269. Wang J, Whiteman MW, Lian H, Wang G, Singh A, Huang D, Denmark T. A non-canonical MEK/ERK signaling pathway regulates autophagy via regulating Beclin 1. J Biol Chem 284: 21412–21424, 2009. doi:10.1074/jbc.M109.026013.
    Crossref | PubMed | ISI | Google Scholar
  • 270. Wang X, Winter D, Ashrafi G, Schlehe J, Wong YL, Selkoe D, Rice S, Steen J, LaVoie MJ, Schwarz TL. PINK1 and Parkin target Miro for phosphorylation and degradation to arrest mitochondrial motility. Cell 147: 893–906, 2011. doi:10.1016/j.cell.2011.10.018.
    Crossref | PubMed | ISI | Google Scholar
  • 271. Wang Y, Nartiss Y, Steipe B, McQuibban GA, Kim PK. ROS-induced mitochondrial depolarization initiates PARK2/PARKIN-dependent mitochondrial degradation by autophagy. Autophagy 8: 1462–1476, 2012. doi:10.4161/auto.21211.
    Crossref | PubMed | ISI | Google Scholar
  • 272. Wei Y, Pattingre S, Sinha S, Bassik M, Levine B. JNK1-mediated phosphorylation of Bcl-2 regulates starvation-induced autophagy. Mol Cell 30: 678–688, 2008. doi:10.1016/j.molcel.2008.06.001.
    Crossref | PubMed | ISI | Google Scholar
  • 273. White E. Deconvoluting the context-dependent role for autophagy in cancer. Nat Rev Cancer 12: 401–410, 2012. doi:10.1038/nrc3262.
    Crossref | PubMed | ISI | Google Scholar
  • 274. White Z, Terrill J, White RB, McMahon C, Sheard P, Grounds MD, Shavlakadze T. Voluntary resistance wheel exercise from mid-life prevents sarcopenia and increases markers of mitochondrial function and autophagy in muscles of old male and female C57BL/6J mice. Skelet Muscle 6: 45, 2016. [Erratum in Skelet Muscle 7: 4, 2017. 10.1186/s13395-017-0120-3. 28202058.] doi:10.1186/s13395-016-0117-3.
    Crossref | PubMed | ISI | Google Scholar
  • 275. Wirawan E, Vande Walle L, Kersse K, Cornelis S, Claerhout S, Vanoverberghe I, Roelandt R, De Rycke R, Verspurten J, Declercq W, Agostinis P, Vanden Berghe T, Lippens S, Vandenabeele P. Caspase-mediated cleavage of Beclin-1 inactivates Beclin-1-induced autophagy and enhances apoptosis by promoting the release of proapoptotic factors from mitochondria. Cell Death Dis 1: e18 , 2010. doi:10.1038/cddis.2009.16.
    Crossref | PubMed | ISI | Google Scholar
  • 276. Wohlgemuth SE, Seo AY, Marzetti E, Lees HA, Leeuwenburgh C. Skeletal muscle autophagy and apoptosis during aging: effects of calorie restriction and life-long exercise. Exp Gerontol 45: 138–148, 2010. doi:10.1016/j.exger.2009.11.002.
    Crossref | PubMed | ISI | Google Scholar
  • 277. Wu Z, Woodring PJ, Bhakta KS, Tamura K, Wen F, Feramisco JR, Karin M, Wang JY, Puri PL. p38 and extracellular signal-regulated kinases regulate the myogenic program at multiple steps. Mol Cell Biol 20: 3951–3964, 2000. doi:10.1128/MCB.20.11.3951-3964.2000.
    Crossref | PubMed | ISI | Google Scholar
  • 278. Xu Y, Yuan J, Lipinski MM. Live imaging and single-cell analysis reveal differential dynamics of autophagy and apoptosis. Autophagy 9: 1418–1430, 2013. doi:10.4161/auto.25080.
    Crossref | PubMed | ISI | Google Scholar
  • 279. Yang Y, Xing D, Zhou F, Chen Q. Mitochondrial autophagy protects against heat shock-induced apoptosis through reducing cytosolic cytochrome c release and downstream caspase-3 activation. Biochem Biophys Res Commun 395: 190–195, 2010. doi:10.1016/j.bbrc.2010.03.155.
    Crossref | PubMed | ISI | Google Scholar
  • 280. Yatim N, Cullen S, Albert ML. Dying cells actively regulate adaptive immune responses. Nat Rev Immunol 17: 262–275, 2017. doi:10.1038/nri.2017.9.
    Crossref | PubMed | ISI | Google Scholar
  • 281. Yingzhong C, Lin C, Chunbin W. Clinical effects of cyclosporine A on reperfusion injury in myocardial infarction: a meta-analysis of randomized controlled trials. Springerplus 5: 1117, 2016. doi:10.1186/s40064-016-2751-y.
    Crossref | PubMed | Google Scholar
  • 282. Yoshii SR, Kishi C, Ishihara N, Mizushima N. Parkin mediates proteasome-dependent protein degradation and rupture of the outer mitochondrial membrane. J Biol Chem 286: 19630–19640, 2011. doi:10.1074/jbc.M110.209338.
    Crossref | PubMed | ISI | Google Scholar
  • 283. Young MM, Takahashi Y, Khan O, Park S, Hori T, Yun J, Sharma AK, Amin S, Hu CD, Zhang J, Kester M, Wang HG. Autophagosomal membrane serves as platform for intracellular death-inducing signaling complex (iDISC)-mediated caspase-8 activation and apoptosis. J Biol Chem 287: 12455–12468, 2012. doi:10.1074/jbc.M111.309104.
    Crossref | PubMed | ISI | Google Scholar
  • 284. Yousefi S, Perozzo R, Schmid I, Ziemiecki A, Schaffner T, Scapozza L, Brunner T, Simon HU. Calpain-mediated cleavage of Atg5 switches autophagy to apoptosis. Nat Cell Biol 8: 1124–1132, 2006. doi:10.1038/ncb1482.
    Crossref | PubMed | ISI | Google Scholar
  • 285. Zhang J, Ney PA. Role of BNIP3 and NIX in cell death, autophagy, and mitophagy. Cell Death Differ 16: 939–946, 2009. doi:10.1038/cdd.2009.16.
    Crossref | PubMed | ISI | Google Scholar
  • 286. Zhang T, Xue L, Li L, Tang C, Wan Z, Wang R, Tan J, Tan Y, Han H, Tian R, Billiar TR, Tao WA, Zhang Z. BNIP3 protein suppresses PINK1 kinase proteolytic cleavage to promote mitophagy. J Biol Chem 291: 21616–21629, 2016. doi:10.1074/jbc.M116.733410.
    Crossref | PubMed | ISI | Google Scholar
  • 287. Zhang XD, Qi L, Wu JC, Qin ZH. DRAM1 regulates autophagy flux through lysosomes. PLoS One 8: e63245 , 2013. doi:10.1371/journal.pone.0063245.
    Crossref | PubMed | ISI | Google Scholar
  • 288. Zhao J, Brault JJ, Schild A, Cao P, Sandri M, Schiaffino S, Lecker SH, Goldberg AL. FoxO3 coordinately activates protein degradation by the autophagic/lysosomal and proteasomal pathways in atrophying muscle cells. Cell Metab 6: 472–483, 2007. doi:10.1016/j.cmet.2007.11.004.
    Crossref | PubMed | ISI | Google Scholar
  • 289. Zheng Y, Shi Y, Tian C, Jiang C, Jin H, Chen J, Almasan A, Tang H, Chen Q. Essential role of the voltage-dependent anion channel (VDAC) in mitochondrial permeability transition pore opening and cytochrome c release induced by arsenic trioxide. Oncogene 23: 1239–1247, 2004. doi:10.1038/sj.onc.1207205.
    Crossref | PubMed | ISI | Google Scholar
  • 290. Zhu Y, Massen S, Terenzio M, Lang V, Chen-Lindner S, Eils R, Novak I, Dikic I, Hamacher-Brady A, Brady NR. Modulation of serines 17 and 24 in the LC3-interacting region of Bnip3 determines pro-survival mitophagy versus apoptosis. J Biol Chem 288: 1099–1113, 2013. doi:10.1074/jbc.M112.399345.
    Crossref | PubMed | ISI | Google Scholar

AUTHOR NOTES

  • Address for reprint requests and other correspondence: J. Quadrilatero, Univ. of Waterloo, 200 University Ave., Waterloo, ON, Canada N2L 3G1 (e-mail: ).