Keywords: apoptosis, cell death, heart disease, necroptosis, necrosis
Abstract
Twelve regulated cell death programs have been described. We review in detail the basic biology of nine including death receptor-mediated apoptosis, death receptor-mediated necrosis (necroptosis), mitochondrial-mediated apoptosis, mitochondrial-mediated necrosis, autophagy-dependent cell death, ferroptosis, pyroptosis, parthanatos, and immunogenic cell death. This is followed by a dissection of the roles of these cell death programs in the major cardiac syndromes: myocardial infarction and heart failure. The most important conclusion relevant to heart disease is that regulated forms of cardiomyocyte death play important roles in both myocardial infarction with reperfusion (ischemia/reperfusion) and heart failure. While a role for apoptosis in ischemia/reperfusion cannot be excluded, regulated forms of necrosis, through both death receptor and mitochondrial pathways, are critical. Ferroptosis and parthanatos are also likely important in ischemia/reperfusion, although it is unclear if these entities are functioning as independent death programs or as amplification mechanisms for necrotic cell death. Pyroptosis may also contribute to ischemia/reperfusion injury, but potentially through effects in non-cardiomyocytes. Cardiomyocyte loss through apoptosis and necrosis is also an important component in the pathogenesis of heart failure and is mediated by both death receptor and mitochondrial signaling. Roles for immunogenic cell death in cardiac disease remain to be defined but merit study in this era of immune checkpoint cancer therapy. Biology-based approaches to inhibit cell death in the various cardiac syndromes are also discussed.
I. CELL DEATH: INTRODUCTION AND HISTORICAL PERSPECTIVE
Prior to the 1980s, cell death was generally considered passive and unregulated, the result of direct damage to cellular components by overwhelming physical, chemical, or biological insults. Even when apoptosis was described in 1972 as a “new” form of cell death that is morphologically distinct from necrosis (235), the notion that it represented a regulated process was not widely considered.
One exception to the concept of unregulated cell death, however, was the elimination of specific cells at specific times during development in invertebrates and vertebrates (153, 175, 294, 422). The term programmed cell death was initially coined to refer specifically to these developmental cell deaths, although this term is now applied more broadly. While the molecular basis for developmental cell death was not understood, the temporospatial consistency with which cells were eliminated in this context suggested an active and regulated process. For example, in the roundworm Caenorhabditis elegans, 131 cells in hermaphrodite embryos and 147 cells in male embryos (out of 1,090 and 1,178 somatic cells generated respectively) die invariantly at specific times and places. In the 1980–90s, Horvitz and colleagues identified a relatively small network of genes that orchestrate these developmental cell deaths: cell death abnormal (ced)-9 → ced-4 → ced-3 (reviewed in Ref. 302). The discovery was profound as it demonstrated unequivocally that cell death in this context was not occurring passively through unregulated damage to cellular components. Rather, these cell deaths were actively mediated through a signaling pathway shown in subsequent genetic mosaic analyses to be operating in a cell-autonomous manner (555). This work established the genetic basis of a regulated and actively mediated cell suicide process.
This same genetic pathway has been conserved over 600 million years of evolution to humans where, importantly, it mediates not only developmental cell deaths, but also cell death in postnatal life. The orthologs of ced-9, ced-4, and ced-3 in mammals encode the Bcl-2 family (187), Apaf-1 (575), and the caspase family (554), respectively, each major components of the mitochondrial apoptosis pathway. In higher organisms, apoptosis is also mediated via a second pathway involving plasma membrane death receptors (9, 14, 142). During the 1990s, intensive research expanded these genetic relationships into the biochemical events that constitute the mitochondrial and death receptor pathways (30, 263, 267, 284, 299, 338, 509).
During this important period of discovery, the prevailing assumption was that apoptosis was the sole form of actively mediated and regulated cell death. Other death processes (e.g., necrosis) were assumed to be passive and unregulated. By the beginning of the new millennium, however, it had become clear that apoptosis was not the only regulated form of cell death. Although necrosis has traditionally been viewed as unregulated, it is now recognized that a significant proportion of necrotic cell deaths, the exact proportion unclear, is actively mediated through regulated pathways. Although the signaling is distinct from apoptosis, these necrosis pathways also involve mitochondria (17) and death receptors, the latter referred to as necroptosis (194, 323, 496). In hindsight, the existence of regulated necrosis pathways in vertebrates should have come as no surprise as forms of regulated necrosis also exist in C. elegans (533).
In addition to apoptosis and regulated necrosis, a number of other death programs have emerged including ferroptosis (97, 456), pyroptosis (128, 438), parthanatos (121, 511), entotic cell death (247, 371), NETotic cell death (136, 142), lysosome-dependent cell death (433), autophagy-dependent cell death (11, 86, 289, 290), and immunogenic cell death (140, 321). While some of these programs are ubiquitous, others appear to operate only in very specialized contexts. Moreover, while each is defined by distinct morphological and/or mechanistic features, further investigation is needed to determine the true independence of some of these programs versus their functioning as “subroutines” within another program.
Herein we review current knowledge pertaining to fundamental mechanisms of regulated cell death and the roles of these death programs in the major cardiac syndromes: myocardial infarction (MI) and heart failure (TABLE 1). Emphasis will be on apoptosis and necrosis, although we will also consider autophagy-dependent cell death, ferroptosis, pyroptosis, parthanatos, and immunogenic cell death. We do not discuss entotic cell death, NETotic cell death, or lysosome-dependent cell death. Thus far, entotic cell death and lysosome-dependent cell death have not been demonstrated to mediate the demise of cardiac cells, and NETotic cell death has been implicated only indirectly through effects on neutrophils (32, 489, 519).
Table 1.
Clinician call-out box
Cell Death in Myocardial Infarction and Heart Failure |
---|
1. Cell death can occur in a passive manner or through multiple actively mediated cell suicide programs referred to collectively as regulated cell death. |
2. Regulated heart muscle cell death plays important roles in the pathogenesis of reperfused myocardial infarction (ischemia/reperfusion) and heart failure with reduced ejection fraction. |
3. Because regulated cell death is actively mediated through molecular pathways, the possibility exists to inhibit this signaling to reduce cardiac damage and dysfunction from these major cardiac syndromes. |
II. MECHANISMS OF APOPTOSIS AND NECROSIS
In this section, we will discuss basic aspects of apoptosis and necrosis signaling in depth. Mechanistic features of autophagy-dependent cell death, ferroptosis, pyroptosis, parthanatos, and immunogenic cell death will be reviewed in section III in parallel with disease implications.
A. Morphological Definitions
Cell death was first characterized by light microscopy. Although not sensitive enough to detect the earliest evidence of irreversible cellular damage (211), this technique is useful in contrasting the morphological characteristics of apoptosis and necrosis. Apoptotic cells are characterized by three cardinal features: 1) shrinkage (apoptosis originally referred to as “shrinkage necrosis”; Refs. 234, 235); 2) fragmentation into membrane-enclosed structures containing mixtures of cell parts (apoptotic bodies); and 3) phagocytosis of apoptotic bodies by macrophages or neighboring cells. Other more variable morphological manifestations of apoptosis include plasma membrane blebbing and chromatin condensation with DNA typically plastered against the inner nuclear membrane, although chromatin condensation can also be observed in necrotic cells. The time frame for cell shrinkage, fragmentation, and phagocytosis varies greatly among organisms and tissues ranging from minutes to hours. When apoptosis proceeds efficiently, apoptotic bodies are removed without leakage of intracellular contents into the extracellular space, thereby avoiding inflammation. Moreover, apoptotic cells actively suppress an immune response through a variety of mechanisms (25, 486). Because of its rapidity and the absence of accompanying tissue inflammation, apoptosis is often a stealth process with single or small numbers of cells disappearing without a trace. In contrast, while cells stimulated to undergo apoptosis in culture usually exhibit typical apoptotic features at early time points, they often acquire necrotic features (e.g., plasma membrane leakiness) at late time points. The latter is referred to as “secondary necrosis” and reflects the absence of a phagocytic cleanup system in vitro.
While apoptosis and regulated necrosis were recognized only ~50 and ~20 yr ago, respectively, the concept of necrotic cell death as a morphological entity dates back more than 150 yr to the pioneering work of Virchow, who referred to this death process as “necrobiosis” (499). Necrotic cells exhibit cell and organelle swelling (due to pan-membrane dysfunction) and loss of defined cellular architecture (404). Necrosis in tissues in vivo typically involves swaths of contiguous (or proximate) cells, rather than single isolated cells as in apoptosis. Additionally, necrosis is accompanied by marked acute and chronic inflammation. The latter is induced by the passive and active release of mediators that recruit inflammatory/immune cells (50, 474). With time, inflammation often transitions to fibrosis including entrapment of remnants of dead cells. These features provide a permanent record that necrotic cell death has occurred.
B. Overview of Apoptosis and Necrosis Signaling
While morphology is informative, a key objective is to understand the various death programs at the molecular level. Both apoptosis and necrosis can be transduced through pathways involving cell surface death receptors (also referred to as the extrinsic pathway) or mitochondria (intrinsic pathway) (FIGURE 1). In the case of apoptosis, both pathways lead to enzymatic activation of caspases, which are a subclass of cysteine-dependent proteases that hydrolyze peptide bonds following aspartic acid residues in certain motifs (390). While the concept of “caspase-independent apoptosis” [e.g., mediated by apoptosis inducing factor (AIF) (462)] was entertained at one point in the evolution of cell death thinking, caspases are now considered obligatory for apoptosis. In contrast, AIF is now believed to have roles in necrosis and parthanatos that will be discussed below (248, 334).
FIGURE 1.
Overview of apoptosis and necrosis pathways. Both apoptosis and necrosis can be mediated through pathways that involve death receptors or mitochondria, resulting in four distinct pathways: 1) death receptor-mediated apoptosis, 2) death receptor-mediated necrosis (necroptosis), 3) mitochondrial-mediated apoptosis, and 4) mitochondrial-mediated necrosis. Regardless of upstream signaling, apoptosis in both pathways leads to activation of the cysteine-dependent proteases, called caspases. In death receptor-mediated apoptosis, binding of death ligands to their cognate receptors results in the formation of sequential protein complexes (complexes I and II). In the latter, caspase-8 (or -10, not shown) is activated. These caspases then cleave and activate downstream caspases, such as -3 and -7. In mitochondrial-mediated apoptosis, caspases are activated in a cytosolic complex (not shown) whose formation is triggered by cytochrome c. Cytochrome c gains access to the cytosol following permeabilization of the outer mitochondrial membrane, which is mediated by B cell lymphoma-2 (BCL-2)-associated X protein (BAX) and BCL-2 antagonist/killer 1 (BAK). In contrast to caspases in apoptosis, a single unifying mechanism has not yet been identified for downstream necrosis signaling that is common to both death receptor and mitochondrial pathways. In necroptosis, activation of the serine/threonine kinases receptor interacting protein kinase 1 (RIPK1) and RIPK3 is critical. RIPK3 then phosphorylates and activates a pseudokinase called mixed lineage kinase-like domain (MLKL), which oligomerizes and permeabilizes the plasma membrane to induce necroptosis. The critical event in the induction of mitochondrial necrosis is Ca2+-triggered opening of the mitochondrial permeability transition pore (mPTP) in the inner mitochondrial membrane. This opening causes rapid dissipation of the proton gradient across the inner membrane that drives ATP synthesis leading to energetic deficits. However, the precise mechanism leading to plasma membrane dysfunction in this pathway is unclear (see text).
Caspases exist in a hierarchy. In both the death receptor and mitochondrial pathways, the most upstream caspases, which are called initiator caspases, acquire enzymatic activity when their zymogen forms (called procaspases) are recruited into specific multiprotein complexes characteristic of each pathway. In the death receptor pathway, these complexes are assembled in response to the binding of ligand to death receptor. In the mitochondrial pathway, the critical event is permeabilization of the outer mitochondrial membrane (OMM), allowing the release of cytochrome c into the cytosol where it functions as a cofactor in assembly of the apoptosome (see below).
In contrast to the role of caspases as the final common pathway in apoptosis, no single molecule has emerged that unifies downstream necrosis signaling in the death receptor and mitochondrial pathways. In the death receptor pathway, the critical event in the induction of necroptosis is the activation of receptor interacting protein kinase 3 (RIPK3), a serine/threonine kinase. RIPK3 is often activated through phosphorylation carried out by the homologous receptor interacting protein kinase 1 (RIPK1) but, as discussed below, this is not always the case as there are alternative mechanisms. RIPK3 then phosphorylates and activates a pseudokinase termed mixed lineage kinase-like domain (MLKL), which oligomerizes and permeabilizes the plasma membrane to bring about necroptosis. RIPK3 → MLKL signaling is considered the canonical signaling module for necroptosis.
In contrast to the necroptosis pathway, the defining event in the mitochondrial necrosis pathway is calcium (Ca2+)-dependent opening of the mitochondrial permeability transition pore (mPTP) located on the inner mitochondrial membrane (IMM). Note that, although significantly more complex, the most critical events in apoptosis and necrosis in the mitochondrial pathway involve the OMM and IMM, respectively. Opening of the mPTP during necrosis rapidly dissipates the proton gradient across the IMM that drives mitochondrial ATP synthesis. It remains unclear, however, exactly how the resulting energetic deficit triggers manifestations of necrosis including loss of plasma membrane integrity. For example, does this result simply from dysfunction of plasma membrane ion pumps and osmotic shifts? Alternatively, are more active mechanisms involved as described above for necroptosis and as will be discussed below for pyroptosis? This issue has not been resolved.
C. Caspases
Because of the centrality of caspases to apoptosis and the complexity of their regulation, we will first describe these proteases in more detail before proceeding to signaling in the death receptor and mitochondrial pathways. Caspases are synthesized as largely inactive pro-enzymes termed procaspases. Each contains a prodomain, p20 subunit, and p10 subunit (390). The prodomain mediates important interactions of the procaspase with other proteins, the p20 subunit contains the catalytic cysteine-histidine dyad, and the p10 subunit contributes to substrate specificity. There are three subclasses of procaspases: 1) initiator (also called upstream, apical, or signaling) procaspases, including human procaspases-8, -9, -10, and perhaps -2 (whose role in cell death remains murky; Ref. 35); 2) effector (also called downstream or executioner) procaspases, including human procaspases-3, -6, and -7; and 3) inflammatory procaspases, including human procaspases-1, -4, -5, and perhaps -12 (although caspase-12 is enzymatically dead in most humans due to genetic variation raising questions about its function; Ref. 413). Most procaspases are expressed ubiquitously, exceptions being procaspase-1, which is most abundant in macrophages and monocytes, and procaspase-14, which is expressed in keratinocytes and whose function is unclear (84, 85). In addition, the substrate recognition motifs of the various caspases differ. While they each hydrolyze peptide bonds following only aspartic acid residues (P1 position), the amino acid 3 position upstream of the aspartic acid (P4 position) is also important for caspase specificity. The P4 residue tends to be a branched chain amino acid for the initiator caspases, a second aspartic acid for the effector caspases (although a valine in the case of caspase-6), and a bulky aromatic amino acid for the inflammatory caspases (355).
Apoptosis is mediated by the initiator and effector procaspases. In contrast, the inflammatory procaspases are involved in inflammasome signaling, cytokine processing, and pyroptosis, the latter a nonapoptotic form of cell death related to inflammasome activation (305, 391, 392). In addition to these roles, some caspases also function in non-cell death roles involving differentiation, proliferation, and other cellular processes (312).
Initiator procaspases exist in healthy cells as inactive monomers. Their activation during apoptosis involves a combination of dimerization and trans cleavage (28, 362), both processes induced by their recruitment into multiprotein complexes. Once activated, initiator caspases activate effector procaspases, which exist as inactive dimers in healthy cells. The activation event for effector caspases is cleavage by initiator caspases. This cutting takes place following aspartic acid residues strategically located at the junctions between prodomain, p20, and p10 subunits. The subsequent noncovalent assembly of two p20 subunits and two p10 subunits results in the active effector caspase.
Effector caspases subsequently cut multiple cellular proteins, structural and signaling, to bring about the apoptotic phenotype (129, 218). For example, caspase-3-mediated cleavage of the scramblase Xkr8 (leading to its activation) (463) and of the flippase ATP11C (leading to its inactivation) (432) are responsible for moving phosphatidylserine motifs to the outer leaflet of the plasma membrane, where they provide “eat me” signals instructing macrophages to phagocytose apoptotic bodies. Other examples include cutting of lamin A by caspase-6 (447) and of acinus (411), helicard (243), and inhibitor of caspase-activated DNase/DNA fragmentation factor 45 kD (ICAD/DFF45) (115, 285, 287, 412) by caspase-3 that bring about nuclear apoptotic changes. These examples aside, the functional significance of many instances of caspase-mediated cleavage of cellular proteins remains incompletely understood.
D. Death Receptor Pathway
1. Activating mechanisms: survival, apoptosis, and necroptosis
This pathway is regulated by the binding of specific ligands to their cognate cell surface receptors. Thus, in contrast to the mitochondrial pathway, which transduces a broad spectrum of death stressors, the death receptor pathway responds to a relatively circumscribed subset of external stimuli. Moreover, while the outcome of death receptor signaling was initially thought to be only apoptosis, it was later recognized that these receptors can also promote necroptosis, cell survival, proliferation, and inflammation (142, 190, 434, 496, 523).
Two classes of death receptors have been recognized. The traditional variety, all members of the tumor necrosis factor (TNF) superfamily, activate cell death following ligand binding (14). In contrast, so-called “dependency receptors,” which are molecularly heterogeneous, transmit death signals only when unliganded (314). Interestingly, some dependency receptors also activate survival and trophic pathways when bound by their ligands. Dependency receptors have been much less studied than the traditional variety of death receptors and remain controversial. Some potential dependency receptors [e.g., tropomyosin-related receptor A (TrkA) (44)] have been studied in the heart with respect to their trophic and survival functions, but not with respect to whether they induce cell death when unliganded (dependency mode).
Traditional death receptors include Fas (also called CD95 or Apo-1), TNF receptor 1 (TNFR1), TNF-related apoptosis inducing ligand (TRAIL) receptor 1 (also known as DR4), and TRAIL receptor 2 (also known as DR5) (277, 369, 445, 455, 500). Fas is bound and activated by Fas ligand (FasL; also known as CD95L and Apo-1L), which is an integral plasma membrane protein on an adjacent cell. Notably, Fas is found on most cells, whereas the basal expression of FasL is restricted to specific cell types (e.g., cells in immunologically privileged tissue contexts) (159, 161). But, FasL expression may be induced in response to stress, including during ischemia/reperfusion (I/R) in the heart (213). In contrast to FasL, TNF-α and TRAIL, the ligands for TNFR1 and TRAIL receptors, respectively, are soluble proteins. Death receptors generally exist as preformed trimers (135, 445). When bound by their ligand, trimers are thought to undergo a conformational change that is transmitted to their intracellular domain. This triggers assembly of one or more intracellular protein complexes at the plasma membrane. These complexes provide the platforms that determine which of the potential outcomes will result from death receptor activation.
Before describing these complexes in detail, some general principles will be noted that govern interactions between the core components of these complexes. The most important interactions are mediated in a modular fashion by death-fold motifs. While highly divergent at the primary amino acid level, death-fold motifs are structurally similar in three dimensions, each consisting of six anti-parallel α-helices (236). There are several varieties of death-fold motifs including death domains (DD), death effector domains (DED), caspase recruitment domains (CARD), and pyrin domains (PYD). Generally, death-fold motifs engage in homotypic interactions (binding to the same type of death-fold motif), although heterotypic death-fold interactions have also been reported (347).
Death receptors contain a DD in their intracellular tail which is revealed following ligand binding to the extracellular domain (373). The DD mediates protein complex formation at the plasma membrane. Two classes of complexes have been described: death inducing signaling complex (DISC) and complex I. The DISC was first discovered and initially thought to mediate all instances of apoptosis induced by death receptors (30, 338). When it became clear that death receptor signaling could also result in other outcomes including necroptosis (194, 496), further investigation led to the discovery of complex I (323). The DISC has been best characterized in Fas signaling (431, 510), while complex I has been most intensively studied with respect to TNFR1 (91). However, the relative usage of the DISC versus complex I in mediating apoptosis has not been established and may be context-dependent.
The DISC consists of death receptor, one or two adaptor proteins, and procaspase-8 or -10 (257). Note that humans possess both procaspases-8 and -10, which are highly homologous, although procaspase-8 seems to be functionally more important (469). Mice express only procaspase-8. Following ligand binding, Fas recruits Fas-associated death domain protein (FADD) through an interaction mediated by the DDs in each protein (55). FADD also contains a DED on its opposite end and uses this to recruit procaspase-8, which contains two DEDs in its prodomain. The recruitment of procaspase-8 triggers its activation through forced proximity and trans cleavage as previously discussed. Caspase-8 goes on to cleave and activate downstream effector procaspases to induce apoptosis.
In contrast to the DISC, signaling through complex I can promote several divergent outcomes: cell survival, proliferation, inflammation, apoptosis, or necroptosis (5, 492). Assembly of complex I begins similarly to the DISC. In the case of TNFR1, however, binding of TNF-α results in the recruitment of a second adaptor protein TNFR1-associated death domain protein (TRADD), containing a DD at each end, to the cytoplasmic tail of TNFR1 via DD-DD interactions (FIGURE 2). Furthermore, rather than recruiting FADD as in the case of the DISC, TRADD recruits RIPK1 (454), also through DD-DD interactions (323). RIPK1 may also be able to bind directly to TNFR1 through DD-DD interactions without the interposition of TRADD. Complex I also includes several other proteins. The adaptor proteins TNF receptor-associated factors 2 and 5 (TRAF2 and TRAF5) and cellular inhibitor of apoptosis proteins 1 and 2 (cIAP1 and 2) are recruited to TRADD and catalyze the attachment of lysine 63 (K63)-linked ubiquitin chains onto RIPK1 (23). [Note that the name “inhibitor of apoptosis proteins” reflects a separate function of cIAP1 and -2 in the mitochondrial pathway, which will be discussed below (87).] K63 ubiquitination facilitates recruitment of the linear ubiquitin chain assembly complex (LUBAC), consisting of heme-oxidized iron responsive element binding protein 2 (IRP2) ubiquitin ligase 1 (HOIL-1; which is the catalytic subunit), HOIL-1 interacting protein (HOIP), and shank-associated RH domain interactor (SHARPIN) (138). LUBAC attaches linear [i.e., methionine 1 (M1)-linked] ubiquitin chains onto RIPK1. Also recruited into this complex are cylindromatosis (CYLD) (105, 244, 485), a deubiquitinase that removes primarily linear ubiquitin chains, and its adaptor protein spermatogenesis-associated 2 (SPATA2) that mediates CYLD recruitment into complex I (113, 249, 426, 521); and A20 (526), a deubiquitinase that removes K63-linked ubiquitin chains (526), and its binding protein A20 binding inhibitor of NF-κB-1 (ABIN-1) (109). Together these enzymes determine the ubiquitination state of RIPK1 and other proteins in complex I (502).
FIGURE 2.
Death receptor apoptosis and necroptosis pathways. Death receptor signaling is initiated by death ligand binding to its specific plasma membrane death receptor. Shown here is the binding of tumor necrosis factor-α (TNFα) to TNF receptor 1 (TNFR1). This initiates assembly of complex I on the cytoplasmic tail of the receptor. Outcomes downstream of complex I assembly include cell survival, apoptosis, or necroptosis. In the formation of complex I, TNFR1 recruits the adaptor protein TNFR1-associated death domain (TRADD) which, in turn, recruits receptor interacting protein kinase 1 (RIPK1). TRADD recruits the adaptor proteins TNF receptor-associated factor 2 and 5 (TRAF2/5) and cellular inhibitor of apoptosis proteins 1 and 2 (cIAP1/2), which attach lysine 63 (K63)-linked ubiquitin chains onto RIPK1. The K63-linked ubiquitin chains promote the recruitment of the linear ubiquitin chain assembly complex (LUBAC), which catalyzes the attachment of linear [or methionine 1 (M1)-linked] ubiquitin chains onto RIPK1. K63-linked chains on RIPK1 promote the activation of transforming growth factor-β-activated kinase 1 (TAK1) through recruitment of TAK1 binding proteins 2 and 3 (TAB2/3). TAK1 [acting through inhibitor of κB kinases (IKKs) and nuclear factor-κB (NF-κB) essential modulator (NEMO) recruited to the linear ubiquitin chains, not shown] promotes the activation of NF-κB. NF-κB is a transcription factor whose targets include multiple genes that promote cell survival and inflammation. In addition, TAK1 also activates mitogen-activated protein kinases (MAPKs), which provide additional survival signals. Cell death initiation requires the transition from complex I to cytosolic complex II. This transition is promoted by decreases in RIPK1 ubiquitination and specifically a decrease in the ratio of linear/K63-linked ubiquitin chains. Cylindromatosis (CYLD) and A20 remove primarily linear- and K63-linked ubiquitin chains, respectively. Complex IIa can take two forms. In one, the binding of TRADD-FADD-procaspase-8 signals RIPK1-independent apoptosis. In the other, binding of activated RIPK1-FADD-procaspase-8 signals RIPK1-dependent apoptosis. Furthermore, caspase-8 inhibits necroptosis by cleaving RIPK1 and RIPK3 (not shown). However, if caspase-8 is inhibited (see text), the binding of activated RIPK1 and RIPK3 in complex IIb leads to RIPK3 activation. RIPK3 then phosphorylates and activates a pseudokinase called mixed lineage kinase-like domain (MLKL), which translocates to and permeabilizes the plasma membrane to cause necroptosis.
The K63-linked ubiquitin chains on RIPK1 facilitate the activation of transforming growth factor-β-activated kinase 1 (TAK1; also called MAPKKK7) through recruitment of TAK1 binding proteins 2 and 3 (TAB2 and TAB3) (37, 223). TAK1 provides cell survival signals through the activation of mitogen-activated protein kinases (MAPKs) and nuclear factor-κB (NF-κB). TAK1-mediated activation of NF-κB is brought about through its phosphorylation of inhibitor of κB kinase α and β (IKKα and -β) which involves the recruitment of these IKKs and NF-κB essential modulator (NEMO; also called IKKγ) to linear ubiquitination chains on RIPK1 (110). Transcriptional targets of NF-κB that mediate cell survival include B cell lymphoma-2 (BCL-2), BCL-2-like-1 long form (BCL-xL), cIAP1 and -2, X-linked inhibitor of apoptosis (XIAP), and cellular-FADD-like interleukin-1β-converting enzyme-inhibitory protein (c-FLIP) (46, 193, 246, 291, 322, 494). Importantly, these pro-survival functions of RIPK1 are mediated independently of its kinase activity.
Although RIPK1 can be bypassed in the induction of apoptosis and necroptosis in the death receptor pathway (discussed below), when RIPK1 mediates either of these death outcomes, its kinase activity is required. Both ubiquitination and phosphorylation play important roles in regulating RIPK1 kinase activity. Decreases in the ratio of linear to K63-linked ubiquitin modifications (e.g., in response to changes in CYLD, SPATA2, A20, ABIN-1) favor RIPK1 activation (109, 521, 525, 526). In contrast, RIPK1 phosphorylation by TAK1, IKKs, TANK binding kinase 1 (TBK1), and MAPK-activated protein kinase 2 (MK2) is generally inhibitory, although nuances exist based on the phosphorylation site and whether phosphorylation is transient or sustained (101, 209, 252, 316, 532). These regulatory mechanisms have recently been reviewed in detail (434).
Apoptosis and necroptosis are mediated by different cytosolic complexes derived from complex I (FIGURE 2). Complex IIa signals apoptosis, while complex IIb directs the cell to undergo necroptosis. Complex IIa can induce apoptosis in either a RIPK1-independent or RIPK1-dependent manner (434). RIPK1-independent apoptosis involves a complex defined by FADD-procaspase-8, which may also contain TRADD (246, 322). In contrast, RIPK1-dependent apoptosis is directed by a complex containing RIPK1-FADD-procaspase-8 (125, 477). Activation of RIPK1 kinase activity in this setting requires the homodimerization of this protein, which is mediated through the DD (315). Moreover, RIPK1 activation is required for its interaction with FADD. The utilization of RIPK1-independent versus RIPK1-dependent apoptosis is influenced by the ubiquitination and phosphorylation states of RIPK1, which regulate kinase activation as discussed above, but the precise mechanisms directing this choice are incompletely understood (102, 150, 209, 521). The final step in the induction of apoptosis in both the RIPK1-independent and RIPK1-dependent modes is the recruitment of procaspase-8 to FADD resulting in active caspase-8 through forced proximity and trans cleavage. Moreover, in addition to signaling apoptosis, caspase-8 activation precludes necroptosis. This is brought about by caspase-8-mediated cleavage of RIPK1 and RIPK3, important mediators of necroptosis (124, 276).
In contrast to apoptosis, necroptosis is mediated by complex IIb (also called the necrosome), which includes RIPK1, RIPK3, and MLKL (FIGURE 2). The binding of TNF-α to TNFR1 induces necroptosis when apoptosis is inhibited. Inhibition of apoptosis may result through various mechanisms. For example, certain viral proteins [e.g., cowpox cytokine response modifier A (CrmA) (394)] inhibit caspase-8. The resulting blockage of apoptosis facilitates viral persistence and amplification. In these situations, the switch to cell killing by necroptosis provides a critical host response to limit viral load (61).
Another factor regulating whether necroptosis takes place is the abundance of c-FLIP (207). The gene encoding c-FLIP gives rise to alternatively spliced transcripts, one encoding c-FLIP short (c-FLIPS) and the other c-FLIP long (c-FLIPL) (39). c-FLIPS consists of only two DEDs similar to those in procaspase-8, while c-FLIPL is homologous to full-length procaspase-8 but contains mutations making it catalytically inactive. Either cFLIP isoform can heterodimerize with procaspase-8, an interaction mediated by their DEDs. Heterodimerization between c-FLIPS and procaspase-8 markedly inhibits procaspase-8 activation (125). In contrast, the effects on caspase-8 activity resulting from heterodimerization between c-FLIPL and procaspase-8 vary with c-FLIPL concentration (200). High c-FLIPL concentrations inhibit caspase-8 activity, but low to equimolar concentrations result in partial activation. At one time, these effects were thought to be mediated by interactions of c-FLIP isoforms with FADD, but recent data suggest that they actually involve direct interactions with procaspase-8 (200). When caspase-8 is inhibited by c-FLIPS or c-FLIPL, apoptosis is blocked and necroptosis results (361). Interestingly, the limited caspase-8 activity provided by procaspase-8-c-FLIP heterodimers inhibits necroptosis, although the mechanism is unclear as these heterodimers appear to cause only limited cleavage of RIPK1 (361). The mechanisms that inhibit apoptosis to promote necroptosis in many physiological and pathophysiological contexts, however, remain to be elucidated.
The kinase activity of RIPK3 is required for necroptosis (61, 183, 277). Although RIPK1 can be bypassed (discussed below), RIPK3 is often activated by the kinase activity of RIPK1. RIPK1 and RIPK3 interact via RIP homotypic interaction motifs (RHIM) resulting in a complex series of phosphorylation events in each protein requiring their kinase activities (61). It has been suggested that autophosphorylation, rather than trans phosphorylation, may be most important (163). In addition, it has recently been shown that the attachment of K63-linked ubiquitin chains to already active RIPK1 by Pellino 1 (PELI1) is required for the RIPK1-RIPK3 interaction and necroptosis (503). Another facilitator of this interaction is the binding of heat shock protein 90 (HSP90) and its co-chaperone CDC37 with RIPK3 (262). Conversely, A20 removes K63-linked ubiquitin chains from RIPK3, which interferes with RIPK1-RIPK3 binding (109).
A major target of RIPK3 in the induction of necroptosis is the pseudokinase MLKL (460, 564). RIPK3-mediated phosphorylation of MLKL exposes a four helical bundle in MLKL that promotes its cysteine-dependent tetramerization, progression to amyloid-like filaments, and translocation to and permeabilization of the plasma membrane (43, 51, 189, 264, 337, 398, 504). Both MLKL phosphorylation and the four cysteines are critical for these events. The small molecule necrosulfonamide, which covalently binds one of the cysteines, inhibits necroptosis downstream of MLKL phosphorylation (460). Conversely, forced polymerization of MLKL permeabilizes the plasma membrane even in the absence of RIPK3 activation (283). The mechanisms by which plasma membrane permeabilization take place and are regulated remain incompletely understood but may involve phosphatidylinositol phosphate binding (100, 104, 504), sodium (Na+) and Ca2+ influx, osmotic changes (43, 51), and activation of the endosomal sorting complexes required for transport (ESCRT)-III machinery (154, 482, 551, 558).
While RIPK1-dependent phosphorylation is critical in the activation of RIPK3 in traditional death receptor-induced necroptosis, RIPK3 and necroptosis can also be activated independently of RIPK1 in other paradigms. For example, in response to pathogen-associated molecular patterns (PAMPs), Toll-like receptors (TLRs) can induce RIPK3 activation through a mechanism that involves Toll-interleukin-1 receptor (TIR) domain-containing adapter-inducing interferon β (TRIF), which, like RIPK1, possesses a RHIM (93, 220). RIPK3 can also be activated independently of RIPK1 by DNA-dependent activator of interferon regulatory factors/Z-DNA binding protein 1 (DAI/ZBP1), another RHIM-containing protein (395, 487, 488). The fact that RIPK3 can be activated independently of RIPK1 has necessitated a change in the definition of the canonical necroptosis signaling module from RIPK1 → RIPK3 → MLKL to the present definition RIPK3 → MLKL.
2. Inhibitory mechanisms revealed by in vivo models
What has been presented thus far is a relatively linear schema in which ligand binding of death receptors promotes cell survival through complex I and induces apoptosis or necroptosis through complexes IIa or IIb, respectively. RIPK1 contributes to the survival outcome independently of its kinase activity. In contrast, while apoptosis and necroptosis can occur without RIPK1, when these forms of cell death are mediated by RIPK1, its kinase activity is required. Finally, necroptosis requires RIPK3 kinase activity and MLKL. While the preceding model describes positive regulation, it lacks some critical negative regulatory mechanisms that were revealed through the study of loss-of-function mutations in mice. Accordingly, we will next review the most critical of these mechanisms. The reader is directed to see TABLE 2 for a more complete list of the mouse models that provided these insights.
Table 2.
Genetic manipulations of death receptor pathway proteins in mice
Genotype | Phenotype | Reference Nos. |
---|---|---|
Ripk3 –/– | Normal development | 354 |
Mlkl –/– | Normal development | 530 |
Ripk1 D138N/D138N | Normal development | 382 |
Ripk1 K45A/K45A | Normal development | 219, 444 |
Ripk1 ΔG26F27/ΔG26F27 | Normal development | 288 |
Procaspase-8 –/– | Lethal E (embryonic day) 10.5 | 495 |
Procaspase-8 –/–; Ripk3 –/– | Normal development | 221, 361 |
Procaspase-8 –/–; Mlkl –/– | Normal development | 8 |
Fadd –/– | Lethal E11.5 | 548 |
Fadd –/–; Ripk3 –/– | Normal development | 8, 92 |
Fadd –/–; Mlkl –/– | Normal development | 8, 508 |
Fadd –/–; Ripk1 –/– | Lethal P (postnatal day) 1–3 | 559 |
Flip –/– | Lethal E10.5 | 547 |
Flip –/–; Ripk3 –/– | Lethal E12 | 92 |
Flip –/–; Ripk3 –/–; Fadd –/– | Normal development | 92 |
Flip –/–; Ripk3 –/–; Procaspase-8 –/– | Normal development | 221 |
Flip –/–; Mlkl –/–; Fadd –/– | Normal development | 8 |
Ripk1 –/– | Lethal P1–3 | 233 |
Ripk1 –/–; Procaspase-8 –/– | Lethal P1–3 | 93, 402 |
Ripk1 –/–; Ripk3 –/– | Lethal P1–3 | 93 |
Ripk1 –/–; Mlkl –/– | Lethal P1–3 | 402 |
Ripk1 –/–; Ripk3 –/–; Fadd –/– | Normal development | 93 |
Ripk1 –/–; Ripk3 –/–; Procaspase-8 –/– | Normal development | 93, 219, 402 |
Ripk1 –/–; Mlkl –/–; Fadd –/– | Normal development | 8 |
Ripk1 –/–; Procaspase-8 –/–; Dai/Zbp1 –/–; Trif –/– | Normal development | 353 |
Ripk1 –/–; Tnfr1 –/– | Lethal P1–3 | 93, 402 |
Ripk1 –/–; Tnfr1 –/–; Trif –/– | Lethal P14–21 | 93 |
Ripk1 –/–; Tnfr1 –/–; Infar –/– | Lethal P14–21 | 93 |
Ripk1 –/–; Tnfr1 –/–; Ripk3 –/– | Normal development | 93 |
Ripk1 mRHIM/mRHIM | Lethal P1–3 | 274, 353 |
Ripk1 mRHIM/mRHIM; Mlkl –/– | Normal development | 274, 353 |
Ripk1 mRHIM/mRHIM; Ripk3–/– | Normal development | 274, 353 |
Ripk1 mRHIM/mRHIM; Dai/Zbp1 –/– | Normal development | 274, 353 |
Ripk1 mRHIM/mRHIM; Trif –/– | Lethal P1–3 | 353 |
Ripk3 D161N/D161N | Lethal E11.5 | 352 |
Ripk3 D161N/D161N; Dai/Zbp1 –/– | Lethal E11.5 | 352 |
Ripk3 D161N/D161N; Cyld –/– | Lethal E11.5 | 352 |
Ripk3 D161N/D161N; Trif –/– | Lethal E11.5 | 352 |
Ripk3 D161N/D161N; Mlkl –/– | Lethal E11.5 | 352 |
Ripk3 D161N/D161N; Tnfr1 –/– | Lethal E11.5 | 352 |
Ripk3 D161N/D161N; Flip –/– | Lethal E11.5 | 352 |
Ripk3 D161N/D161N; Ripk1 –/– | Lethal P1–3 | 352 |
Ripk3 D161N/D161N; Procaspase-8 –/– | Normal development | 352 |
Ripk3 K51A/K51A | Normal development | 303 |
Ripk3 K51A/K51A; Procaspase-8 –/– | Normal development | 303 |
a) negative regulation of apoptosis and necroptosis by ripk1.
Germline deletion of RIPK1 was observed to result in neonatal lethality accompanied by the unleashing of cell death in multiple tissue compartments (233). Subsequent conditional RIPK1 knockouts in various tissues (intestine, skin, blood, and liver) also resulted in cell death at baseline or in response to stress (74, 405, 471, 493). The type of cell death was tissue-dependent with, for example, apoptosis predominating in intestine and necroptosis in skin. Importantly, the consequences of germline RIPK1 deletion could be rescued by simultaneous deletion of 1) FADD or procaspase-8 or TNFR1 (apoptosis arm) and 2) RIPK3 or MLKL or the combination of TRIF and DAI/ZBP1 (necroptosis arm), but not by inactivation of genes in either arm alone (8, 93, 219, 353, 402, 559). In aggregate, these data indicate that the net effect of RIPK1 is to suppress apoptosis and necroptosis.
As described above, RIPK1 may promote cell survival in a kinase-independent manner or cell death (apoptosis or necroptosis) through its kinase activity. To test in vivo why RIPK1 loss was derepressing apoptosis and necroptosis, mice were created with germline knockin of kinase-dead RIPK1. These mice were normal, indicating that the non-kinase function of RIPK1 suppresses cell death in vivo (219, 288, 382, 444). Further investigation showed that knockin of a mutation that disables the RIPK1 RHIM (which mediates RIPK1 interactions with RIPK3) phenocopies the neonatal lethality of RIPK1 absence and can be rescued by deletion of RIPK3, MLKL, or DAI/ZBP1 (necroptosis arm) (274, 353). Taken together, these observations lead to the conclusion that RIPK1 suppresses both apoptosis and necroptosis in the death receptor pathway through a kinase-independent, RHIM-dependent function. Some of the kinase-independent mechanisms by which RIPK1 mediates cell survival in complex I have already been discussed above, and others may operate in complexes I and II.
Interestingly, in contrast to mice lacking RIPK1, four humans were recently reported with homozygous loss-of-function mutations in both RIPK1 alleles (protein absent in 3 of 4 individuals) (68). While these people survived neonatal life, all four manifest lymphopenia, susceptibility to infections, arthritis, or inflammatory bowel disease. In addition, cells derived from these individuals exhibited increased susceptibility to necroptosis.
b) negative regulation of necroptosis by catalytically active c-flipL-procaspase-8 heterodimers.
This mechanism was discussed earlier and originates from the curious observation that germline deletion of FADD or procaspase-8 causes embryonic lethality (495, 548). Why loss of apoptosis signaling was killing the embryo went unexplained for multiple years and led to the incorrect hypothesis that accumulation of excessive cells was responsible. A mechanism was not elucidated until it was observed that simultaneous deletion of RIPK3 or knockin of either of two kinase-dead RIPK3 mutants rescued the lethality of either FADD or procaspase-8 deletion (8, 92, 221, 303, 352, 361). This indicates that necroptosis is activated in the absence of FADD or procaspase-8. Further work, described above, revealed that c-FLIPL-procaspse-8 heterodimers possessing low, but detectable, caspase-8 activity suppressed necroptosis, although the precise substrates remain unclear.
c) as yet unresolved: possible negative regulation of apoptosis by ripk3.
A somewhat surprising finding regarding the regulation of apoptosis emerged from the study of kinase-dead RIPK3 knockin mice. One of these mutants (D161N) exhibited embryonic lethality due to excessive apoptosis in the yolk sac vasculature. This was rescued by simultaneous deletion of procaspase-8 (352). In contrast, no rescue resulted from co-deletion of TNFR1, CYLD, FLIP, TRIF, DAI/ZBP1, or MLKL. The mechanism of this effect is thought to involve complex formation between RIPK3 (D161N), RIPK1, FADD, and procaspase-8 resulting in activation of procaspase-8. In contrast, wild-type RIPK3 does not engage in these interactions. However, a second RIPK3 kinase-dead (K51A) knockin mouse did not exhibit embryonic lethality or activation of apoptosis (303), despite the fact that both RIPK3 kinase-dead mutants were able to rescue embryonic lethality in mice lacking procaspase-8. The differential abilities of the two RIPK3 kinase-dead mutants to activate apoptosis is hypothesized to result from differences in the conformations of RIPK3 resulting from the precise mutations employed rather than from the absence of kinase activity. Further work will be needed to test this hypothesis and its physiological significance.
The conclusions from these in vivo models can be summarized as follows: 1) RIPK1 suppresses early postnatal lethality in mice by inhibiting both apoptosis and necroptosis in the death receptor pathway, a function that is independent of its kinase activity but depends on its RHIM. 2) Caspase activity resulting from the c-FLIPL-procaspase-8 heterodimer suppresses embryonic lethality from FADD or procaspase-8 deletion by inhibiting RIPK3-dependent necroptosis.
E. Mitochondrial Pathway
The mitochondrial pathway, which can signal cell death through apoptosis or necrosis, is activated by diverse stimuli including loss of nutritional/growth/survival factors, hypoxia, ischemia, hypoxia/reoxygenation, ischemia/reperfusion, oxidative stress, nitrosative stress, proteotoxic stress, DNA damage, increases in concentrations of cytoplasmic or mitochondrial Ca2+, and multiple toxins and drugs (e.g., chemotherapeutics and targeted cancer therapies) (73). These stimuli are transduced to mitochondria directly or through other organelles [e.g., endo/sarcoplasmic reticulum (ER/SR)] to bring about cell killing.
The central event in apoptosis in the mitochondrial pathway is permeabilization of the OMM [often referred to as mitochondrial outer membrane permeabilization (MOMP)]. This allows various mitochondrial apoptogens to access the cytosol where they promote procaspase activation. In contrast, the major event in necrosis in the mitochondrial pathway is opening of the mPTP in the IMM (341).
1. Mitochondrial apoptosis pathway
MOMP is regulated by the BCL-2 family proteins (57) (FIGURE 3). There are three subfamilies, which are defined by whether they promote or inhibit cell death and which BCL-2 homology domains (BH) they contain. The first subfamily comprises the pro-survival BCL-2 proteins, which contain BH1–4. These include BCL-2 itself, BCL-xL, myeloid cell leukemia-1 (MCL-1), BCL-2-like protein 2 (BCL-W), and others. The second subfamily is made up of the multi-domain pro-cell death BCL-2 proteins, whose structures are classically defined as containing BH1–3 but, in some cases, have been interpreted to also include BH4. These include BCL-2-associated X protein (BAX) and BCL-2 antagonist/killer 1 (BAK). A third member, BCL-2 related ovarian killer (BOK), is important in embryonic development, but its role in cell death is not clear (232, 293). The third subfamily comprises BH3-only proteins, each of which promotes cell death and contains only the BH3 domain. These include BCL-2-like 11 (BIM), BH3 interacting domain death agonist (BID), p53-upregulated modulator of apoptosis (PUMA), phorbol-12-myristate-13-acetate-induced protein 1 (NOXA), BCL-2-associated agonist of cell death (BAD), BCL-2 interacting killer (BIK), Harakiri (HRK)/BCL-2 interacting protein death protein 5 (HRK/DP5), BCL-2 modifying factor (BMF), and others. These three subfamilies of BCL-2 proteins engage in complex interactions to regulate MOMP. The overview is that BH3-only proteins transduce death signals from the periphery to activate BAX and BAK and inactivate the pro-survival BCL-2 proteins. Conversely, the pro-survival BCL-2 proteins seek to inactivate BAX and BAK and to neutralize the BH3-only proteins.
FIGURE 3.
Mitochondrial apoptosis pathway. The mitochondrial apoptosis pathway can be triggered by diverse extracellular and intracellular stress stimuli. The central event in the execution of this pathway is permeabilization of the outer mitochondrial membrane (OMM), which is tightly regulated by the B cell lymphoma-2 (BCL-2) family proteins. These are divided into three subfamilies according to their function and BCL-2 homology (BH) domains: 1) pro-survival proteins, including BCL-2, BCL-2-like-1 long form (BCL-xL), and myeloid cell leukemia-1 (MCL-1); 2) multidomain pro-cell death proteins, including BCL-2-associated X protein (BAX) and BCL-2 antagonist/killer 1 (BAK); and 3) pro-cell death BH3-only proteins, which are further subdivided into “activators” and “sensitizers.” Activator BH3-only proteins [BCL-2-interacting domain death agonist (BID), BCL-2-interacting mediator of cell death (BIM), p53 upregulated modulator of apoptosis (PUMA), and perhaps phorbol-12-myristate-13-acetate-induced protein 1 (NOXA)] directly bind and conformationally activate BAX and BAK. Conversely, the pro-survival BCL-2 proteins oppose these events both through sequestering the activator BH3-only proteins and through interacting with and inhibiting BAX and BAK. Sensitizer BH3-only proteins [BCL-2 antagonist of cell death (BAD), BCL-2-interacting killer (BIK), BCL-2-modifying factor (BMF), and Harakiri (HRK)] indirectly activate BAX and BAK through binding the pro-survival BCL-2 proteins, which both displaces the activator BH3-only proteins as well as prevents the pro-survival proteins from binding and neutralizing BAX and BAK. Activation of BAX and BAK drives their homo- and hetero-oligomerization within the OMM. This results in membrane permeabilization and release of apoptogenic factors from mitochondria, including cytochrome c, second mitochondria-derived activator of caspase (SMAC)/direct inhibitor of apoptosis (IAP) binding protein with low PI (DIABLO), and OMI/high temperature requirement protein A2 (OMI/HtrA2). The binding of cytochrome c and dATP to Apaf-1 promotes formation of the apoptosome, in which procaspase-9 is activated. Caspase-9 subsequently activates downstream procaspases including -3 and -7. IAPs bind and inhibit procaspase-9 and active caspases-3 and -7. SMAC/DIABLO and OMI/HtrA2 bind and inhibit IAPs resulting in activation of caspases. Caspase-8 can also cleave and activate BID (tBID), allowing tBID to bind and activate BAX, thereby linking the death receptor and mitochondrial apoptosis pathways.
Here we consider this regulation in more detail. A specific subset of BH3-only proteins termed “activators” (which include BID, BIM, PUMA, and perhaps NOXA) bind directly to and induce conformational changes in BAX and BAK (FIGURE 3). The regulation of BAX has been studied in most detail and will be described. While BAX is also regulated transcriptionally (e.g., by p53) (325) and through posttranslational modifications such as phosphorylation (292), its conformational activation through interactions with BH3-only activators is critical. This involves binding of the BH3 helix of a BH3-only activator to the BAX trigger site located in its NH2 terminus (148). This results in displacement of the linker between α-helices 1 and 2 (α1 and α2), rearrangement of internal helices, exposure of BAX’s own BH3 helix, and ultimately exposure of a transmembrane domain in α9, the most COOH-terminal helix (147). This transmembrane domain inserts tightly into the OMM, shifting the location of BAX from the cytoplasm, where it normally resides in healthy cells in an inactive state, to mitochondria. In addition, BAX mitochondrial translocation may also be facilitated by other active mechanisms that are not completely understood (383, 531). While BAX activation begins in the cytoplasm, lipid and protein interactions at the mitochondria also contribute (56). BAK resides constitutively at the OMM, and it also undergoes conformational activation. Following activation, BAX and BAK engage in homo- and hetero-oligomerization within the OMM to induce MOMP. The precise mechanism of MOMP is not clear, but may involve pore formation as suggested by liposome studies. MOMP, in turn, allows release of mitochondrial apoptogens to the cytosol where they facilitate procaspase activation.
The pro-survival BCL-2 proteins inhibit MOMP through several mechanisms. First, they bind and sequester the BH3-only activators, an interaction involving the BH3 domain of the activator and a hydrophobic groove formed by the convergence of BH1–3 in the COOH terminus of the pro-survival BCL-2 protein (referred to hereafter as the “groove”). An additional more controversial mechanism is direct binding of the pro-survival BCL-2 proteins to BAX and BAK, an interaction mediated by the BH3 domain of BAX or BAK and the groove in the pro-survival BCL-2 protein. This inhibitory mechanism is likely applicable only to partially activated BAX and BAK because the BH3 domain needs to be exposed. A third mechanism by which the pro-survival BCL-2 protein BCL-xL inhibits BAX is through promoting BAX retrotranslocation from mitochondria (111).
The BH3-only activators not only activate BAX and BAK through direct binding, but also indirectly by inhibiting the pro-survival BCL-2 proteins. The latter involves binding of the BH3 helix of the activator to the groove in the pro-survival BCL-2 proteins. The remaining BH3-only proteins that are not direct activators of BAX and BAK are called “sensitizers.” Using their BH3 helices, they also can bind the groove in the pro-survival BCL-2 proteins, which will displace the activator BH3-only proteins, releasing them to activate BAX and BAK. The result of this competition reflects the relative stoichiometry and binding constants of the proteins. But, under conditions of apoptosis induction, the sensitizers win out freeing the activators from sequestration.
BH3-only proteins (both activators and sensitizers) become activated through distinct biochemical mechanisms providing a means for them to relay a diversity of death stimuli to BAX and BAK and/or the pro-survival BCL-2 proteins. For example, PUMA and NOXA are transcriptionally activated by p53 (345, 364). BIM is activated by ER stress through mechanisms that increase its abundance. These include BIM dephosphorylation which prevents its ubiquitination and proteasomal degradation, and C/EBP homologous protein (CHOP)-mediated increases in Bim transcription (384). Caspase-8, which is activated during death receptor-mediated apoptosis, cleaves BID to produce truncated BID (tBID), in which the BH3 domain is exposed (164, 263), allowing tBID to then bind and activate BAX and BAK. This mechanism serves to link the death receptor and mitochondrial apoptosis pathways, thereby providing a critical amplification loop needed for death receptor ligands to maximally activate caspases in many cell types. BAD is activated when released from sequestration by 14-3-3 proteins. This results from BAD dephosphorylation at a critical serine residue, which can be triggered by loss of survival factors, such as insulin-like growth factor-1 (IGF-1) and interleukin-3 (IL-3), that reduce AKT activity, or by calcineurin-mediated dephosphorylation (76, 83, 506).
Induction of MOMP by activated BAX and BAK oligomers leads to the release of mitochondrial proteins including cytochrome c, second mitochondria-derived activator of caspase (SMAC)/direct inhibitor of apoptosis (IAP) binding protein with low PI (DIABLO) (106, 497), and OMI/high temperature requirement protein A2 (HtrA2) (118, 464) (FIGURE 3). In the cytosol, these proteins facilitate caspase activation and apoptosis as described below. Also released are AIF (462), which, as previously mentioned, likely plays roles in necrosis and/or parthanatos rather than apoptosis (248, 334), and endonuclease G (EndoG) (266), which was once thought to partner with AIF to cut DNA, but whose role in cell death is currently unclear (112). AIF will be discussed in later sections of this review.
In the cytosol, cytochrome c induces caspase activation by triggering the assembly of a multiprotein complex called the apoptosome (267, 284, 575). This consists of a circular array of seven molecules of the adaptor protein Apaf-1, each bound by one cytochrome c molecule (derived from the mitochondria) and one dATP molecule (from the cytosol), and seven molecules of procaspase-9 (2). The binding of cytochrome c presumably induces a conformational change in Apaf-1 that allows it to interact with procaspase-9 through CARDs in each protein. This induces procaspase-9 activation through forced proximity and trans cleavage, and caspase-9 subsequently cleaves and activates effector caspases.
In the cytosol, various IAP proteins (cIAP1, cIAP2, and XIAP) function as caspase inhibitors (87, 89, 108, 407) (FIGURE 3). These are the same proteins discussed previously in the context of their RIPK1 ubiquitination in complex I. In the mitochondrial pathway, these molecules function to inhibit caspases through several mechanisms, as delineated for XIAP. These include binding to procaspase-9 to prevent its dimerization and activation (442) as well as inhibition of already activated effector caspases-3 and -7 through binding that blocks their substrate sites (47, 199, 403, 465). In addition, XIAP promotes the degradation of effector caspases in the proteasome through its E3-ligase function (466). While these mechanisms inhibit apoptosis, XIAP also promotes its own degradation, presumably to provide a negative-feedback control mechanism (424, 542).
When released in the cytosol during apoptosis, SMAC/DIABLO and OMI/HtrA2 relieve inhibition of caspases by IAPs to allow apoptosis to proceed (464) (FIGURE 3). The mechanism involves binding of SMAC/DIABLO or OMI/HtrA2 to IAPs at or near their caspase binding sites, thereby freeing the caspases (185, 453). In addition, OMI/HtrA2 possesses a serine protease activity that permanently inactivates XIAP through cleavage (118, 464, 537).
Our discussion of the mitochondrial pathway thus far has focused on apoptosis and, more specifically, how BCL-2 proteins link death signals with MOMP and how mitochondrial apoptogens released during MOMP induce caspase activation and cell death. However, mitochondria have long been known to play critical roles in necrotic cell death, dating back to before it was even realized that necrosis could be a regulated process. Accordingly, we will next consider how regulated necrosis is mediated in the mitochondrial pathway and how necrosis and apoptosis signaling at this organelle may interconnect.
2. Mitochondrial necrosis pathway
The details of mitochondrial-mediated necrosis are less well defined than those of apoptosis or necroptosis. Nevertheless, some key principles have emerged. First, the triggering event for necrosis in the mitochondrial pathway is Ca2+-induced opening of the mPTP in the IMM. This occurs quickly, usually within minutes following elevation in Ca2+ concentration within the mitochondrial matrix (for example, see Ref. 528). Importantly, mPTP opening takes place in the absence of, or hours before, MOMP, indicating that mPTP-mediated cell death is not occurring secondarily to mitochondrial-mediated apoptosis. (The mechanism by which cytochrome c release may sometimes occur at late time points following mPTP opening is discussed below.) While a definitive knowledge of the components of mPTP would be needed to test directly the hypothesis that opening of this pore triggers mitochondrial-mediated necrosis, strong evidence is provided by studies in which ppif, the gene encoding cyclophilin D, was deleted in the mouse (17, 341, 425). Cyclophilin D is a peptidyl prolyl cis-trans isomerase that is located in the mitochondrial matrix (66, 172). While not a core component of mPTP, cyclophilin D is a strong positive regulator of Ca2+-induced pore opening (17). Moreover, promotion of mPTP opening by cyclophilin D is dependent on its prolyl isomerase activity, although the relevant substrates have not been identified. Importantly, cells lacking cyclophilin D from various tissues of ppif −/− mice are resistant to Ca2+-induced mPTP opening and necrotic cell death. These data suggest that Ca2+-induced mPTP opening is critical for necrosis in the mitochondrial pathway.
While observations that high Ca2+ concentrations induce mitochondrial swelling date back >60 yr (204, 389), the original definition of mPTP was based on pharmacological criteria (173). mPTP is a nonselective pore permeable to molecules <1.5 kDa in mass, whose opening is promoted by high Ca2+ concentrations in the mitochondrial matrix. Ca2+-induced mPTP opening is potentiated by oxidative stress, increases in phosphate, and depletion of adenine nucleotides (ATP and ADP) and inhibited by high concentrations of protons (H+). Some of these conditions occur during the reperfusion phase of I/R, in particular the oxidative stress and reversal of acidosis. Additional operational specificity was imparted to this pharmacological definition by the observation that cyclosporine A (CsA), an inhibitor of cyclophilin D, inhibits Ca2+-induced mPTP opening (67). As will be discussed in section III, CsA blocks I/R injury in some preclinical models of I/R (273, 367), although it appears to have failed in clinical trials (69, 278, 381).
As recently reviewed (226), most models of mPTP between ~1990–2007 depicted it as complex involving the adenine translocase (ANT) and sometimes the mitochondrial phosphate carrier (PiC) in the IMM, the voltage-dependent anion channel (VDAC) in the OMM, and cyclophilin D in the matrix. ANT, which was often hypothesized to be the pore-forming unit in the IMM, is in fact capable of forming pores in artificial membranes (409). Additionally, bongkrekic acid and atractyloside, which bind and regulate ANT in cells, inhibit or promote mPTP opening, respectively (179, 203). These data notwithstanding, subsequent knockout/knockdown studies have ruled out each of these components as essential to the pore function of mPTP (17, 171, 251, 341). It must be emphasized, however, that despite being excluded as components of the pore, ANT, PiC, and cyclophilin D are each critical sensitizers of Ca2+-induced mPTP opening (17, 239, 251, 341).
If ANT, PiC, VDAC, and cyclophilin D do not form the pore, what does? Unexpectedly, recent work has suggested that components of the F1-F0 ATP synthase in mitochondrial complex V constitute the pore-forming unit of mPTP (22). However, different groups have implicated different sets of subunits including the oligomycin sensitivity-conferring protein (OSCP) in the peripheral stalk of the F1 subunit (152) and the c-subunit in the F0 portion (6, 33). Furthermore, based on reconstitution studies, the group supporting the OSCP model proposed that the pore is composed of dimers of the F1-F0 ATP synthase. Both the OSCP and c-subunit models are supported by RNAi-mediated depletion of OSCP and ATP5G1 and ATP5G3 (the latter two proteins isoforms of the c-subunit). However, other research subsequently challenged these models through loss-of-function experiments using CRISPR/Cas9-mediated gene editing of Atp5O (encoding OSCP), Atp5g1, Atp5g2, and Atp5g3 (all encoding the c-subunit), as well as Atp5f1 (encoding the membrane domain of the b-subunit, which imparts structure to the F1 stalk). Those data report that decreases in these subunits do not result in loss of mPTP function (181, 182). In addition, in silico modeling of the c-subunit has raised doubts as to whether its predicted structural and biophysical properties are consistent with mPTP (569). It should be noted that manipulations of some of these F1-F0 ATP synthase components perturb other aspects of mitochondrial function thereby introducing potential confounding factors. Furthermore, it has been counter-argued that careful examination of the data claiming that deletion of the genes encoding OSCP and the b-subunit does not abolish mPTP function (181) actually demonstrates partial inhibition, supporting the notion that these components of the F1-F0 ATP synthase do contribute to the pore (21). Thus this controversy remains unresolved (16), and further work will be needed to settle this critical issue. In addition, if components of the F1-F0 ATP synthase, in fact, do constitute the pore, a second important issue will be to find mutations that allow a separation of ATP synthetic function from necrosis function (if possible at all), a challenge because both processes involve dissipation of the proton gradient across the IMM.
Irrespective of the identity of mPTP, high Ca2+ concentrations in the mitochondrial matrix are the major stimulus for mPTP opening. Accordingly, an important challenge is to understand how pathophysiological stimuli relevant to various disease scenarios bring about these increases. A plausible pathway to explain this has been suggested for I/R (336) (FIGURE 4). The production of lactic acid in the context of anaerobic metabolism during ischemia creates an intracellular acidosis (64). This is partially corrected by activation of the plasma membrane Na+/H+ exchanger (NHE), which operates in reverse mode to transport H+ out of the cell in exchange for Na+ into the cell (335). The subsequent overload of intracellular Na+ activates the plasma membrane Na+/Ca2+ exchanger (NCX) to eject Na+ at the expense of importing Ca2+ (162, 205). The subsequent rise in cytoplasmic Ca2+ concentration triggers the release of large amounts of Ca2+ from the ER/SR through ryanodine and inositol trisphosphate (IP3) receptors. This Ca2+ reaches the mitochondrial matrix, its transport across the OMM and IMM regulated by VDACs (443) and the mitochondrial Ca2+ uniporter (MCU) (375), respectively. As previously noted, mPTP opening occurs mainly during reperfusion when reactive oxygen species (ROS) and correction of intracellular acidosis sensitize mPTP opening. This schema was the conceptual basis for the use of Na+/H+ exchange inhibitors (e.g., cariporide) in I/R, which showed promise in animal studies but failed in clinical trials for reasons that are not completely clear, perhaps circumvention by redundant mechanisms (31, 228, 335, 478).
FIGURE 4.
Mitochondrial necrosis pathway. The mitochondrial necrosis pathway can be triggered by diverse stimuli including loss of nutrient/survival factors, increase in intracellular calcium (Ca2+), reactive oxygen species (ROS), ischemia, and ischemia/reperfusion. The central event in the execution of this pathway is opening of the mitochondrial permeability transition pore (mPTP) in the inner mitochondrial membrane (IMM). Ischemia deprives the myocardium of oxygen (O2), resulting in anaerobic metabolism and intracellular acidosis. In response, the cell pumps out the excess proton (H+) and accumulates sodium (Na+) by reverse operation of the Na+/H+ ion exchanger (NHE). The excess Na+ is, in turn, pumped out by the Na+/Ca2+ exchanger (NCX), which leads to an increase in intracellular Ca2+. Further increases in Ca2+ levels result from Ca2+-induced Ca2+ release from the endo/sarcoplasmic reticulum (ER/SR), resulting in intracellular Ca2+ overload. Increased concentrations of Ca2+ in the mitochondrial matrix promote mPTP opening during reperfusion which neutralizes intracellular acidosis and stimulates ROS production. The pro-cell death protein BCL-2-associated X protein (BAX) also sensitizes Ca2+-induced mPTP opening and necrosis. The consequences of mPTP opening are 1) loss of the H+ gradient across the IMM (mitochondrial membrane potential, Δψm), which leads to cessation of ATP synthesis, and 2) influx of water into the hyperosmolar mitochondrial matrix leading to mitochondrial swelling and eventual rupture of the outer mitochondrial membrane (OMM).
Multiple signaling molecules have been reported to impact Ca2+-induced mPTP opening (reviewed in Ref. 248), but mechanistic insights are unavailable in most cases. Accordingly, we will comment only on some recent advances. Unexpectedly, the multidomain pro-cell death proteins BAX and BAK have been demonstrated to facilitate Ca2+-induced mPTP opening and necrotic cell death (225, 528). While the details pertaining to heart disease will be discussed in more detail in section III, the basic observation is that mice with combined cardiomyocyte-specific deletion of Bax and germline deletion of Bak exhibit rescue of I/R-induced necrotic morphology (528). Furthermore, absence of BAX and BAK was found to render mouse embryonic fibroblasts (MEFs) resistant to very early Ca2+-induced mPTP opening that precedes mitochondrial cytochrome c release and caspase activation by hours (225, 528). Absence of BAX alone is sufficient for this effect as reintroduction of physiological levels of BAX into Bax/Bak double knockout cells restored sensitivity to Ca2+-induced mPTP opening. The sufficiency of BAK alone has not yet been tested. BAX oligomerization is known to be required for MOMP during apoptosis (151, 238). In contrast, BAX does not undergo oligomerization during necrosis (528). Moreover, oligomerization-defective BAX mutants, which are unable to support MOMP and apoptosis (151, 238), were sufficient to restore sensitization of Ca2+-induced mPTP opening in Bax/Bak double knockout cells (225, 528), providing strong support for the notion that BAX mediates necrosis and apoptosis through fundamentally different mechanisms. The underlying molecular mechanism, however, remains incompletely understood. One group suggests that BAX functions as an OMM component of mPTP by facilitating OMM permeability and conductance (225). Another group proposes that BAX-mediated mitochondrial fusion, a recognized non-cell death function of BAX (196, 224), is important in the sensitization to Ca2+-induced mPTP opening (528). These hypotheses are not mutually exclusive.
In general, the role of mitochondrial dynamics (fusion and fission) in cell death remains poorly defined (306). Multiple studies demonstrate that mitochondria undergo fission during apoptosis, and fission has been hypothesized to facilitate MOMP. MOMP per se, however, can be dissociated from mitochondrial fission by the overexpression of pro-survival BCL-2 proteins, which inhibit MOMP but not fission (436). Nevertheless, mitochondrial fission appears to contribute to apoptosis and MOMP as evidenced by experiments showing that these processes are inhibited by pharmacological or genetic inhibition of dynamin related protein 1 (DRP1), which mediates fission (45, 132). Given these observations, the data showing that increased mitochondrial “connectivity,” whether through BAX-mediated mitochondrial fusion or inhibition of DRP1-mediated fission, sensitizes cells to Ca2+-induced mPTP opening are intriguing (528). In keeping with these observations, genetic deletion of mitofusins 1 and 2 (MFN 1 and 2), which mediate mitochondrial fusion, lessen infarct size during myocardial I/R, which is primarily a necrotic process (174). Somewhat confusing, however, is that pharmacological inhibition of DRP1 has also been reported to reduce infarct size (368). How do we reconcile these data? Do these observations reflect other unrelated functions of the molecules that were manipulated which happen to track with changes in mitochondrial connectivity? For example, DRP1 also mediates BAX oligomerization (329), and MFN2 also functions as a Parkin receptor in mitophagy (52) and in promoting mitochondrial-ER tethering (79, 349). Or do these findings reflect the influence of changes in mitochondrial size and shape on other aspects of mitochondrial anatomy (e.g., cristae structure; Refs. 63, 429) and function? Or, perhaps they reflect fundamental differences in how apoptosis and necrosis are signaled in the mitochondrial pathway? Taken together, these data suggest that mitochondrial dynamics and mitochondrial-mediated cell death (apoptosis and necrosis) are functionally and mechanistically interconnected, but additional work will be required to understand the precise interrelationships.
In thinking about the effects of BAX and BAK even more broadly, it should be noted that, in addition to their actions at the mitochondria, these proteins exert effects at other organelles that could impact mitochondrial-mediated necrosis indirectly. For example, BAX and BAK also reside at the ER (574), where they promote accumulation of luminal Ca2+ through a mechanism that may involve antagonism of a Ca2+ leak induced by the interaction of pro-survival BCL-2 proteins with IP3 receptor 1 (IP3R1) (359, 360). This ensures that adequate reserves of ER Ca2+ are available to respond to a subsequent death stimulus. Moreover, ER-localized BAX may be particularly important in mediating cell death in response to oxidative stress (430). BAX and BAK also induce permeabilization of the lysosomal membrane (227), although the relevance of this to necrotic cell death is not known.
The mechanisms by which mPTP opening brings about necrotic cell death are not clear. Opening of this pore has two major consequences: 1) immediate dissipation of the proton gradient across the IMM leading to cessation of ATP synthesis, and 2) the rapid ingress of water down its osmotic gradient into the solute-rich mitochondrial matrix resulting in mitochondrial swelling. Apoptotic cells shut down many energy-requiring cellular processes, including DNA repair, protein translation, and proteasomal function, often through caspase-mediated cleavage of the relevant effectors. In contrast, necrotic cells not only stop making ATP, but continue to spend it in an unrestrained manner (260, 410, 449, 461). Thus cellular ATP levels plummet during necrosis, a hallmark of this cell death process. This “energetic crisis” is often invoked as the explanation for the loss of plasma membrane integrity, another defining feature of necrotic cells. While deficits in ATP may provoke osmotic stresses through dysfunction of plasma membrane pumps (117), it remains unclear whether this mechanism accounts for plasma membrane leakiness during mitochondrial-mediated necrosis. Other possibilities meriting further exploration include 1) the extent to which loss of ATP disrupts plasma membrane repair processes (27, 498) and 2) the involvement of yet to be identified mechanisms that actively disrupt the plasma membrane, analogous to the roles of MLKL in necroptosis (43, 51, 189, 264, 337, 398, 504) and gasdermins in pyroptosis (184, 230, 406, 439, 512) (discussed in sect. IIIC3).
Some signaling loops appear to operate during necrosis to amplify energy deficits. For example, poly(ADP-ribose) (PAR) polymerase-1 (PARP-1) participates in DNA repair and transcription (245), processes that continue during necrosis and may be further induced (e.g., by oxidative damage to DNA during reperfusion). The synthesis of PAR and its transfer onto target proteins by PARP-1 requires NAD+. Since NAD+ is an essential cofactor for glyceraldehyde-3-phosphate dehydrogenase (GAPDH) in glycolysis, this process is compromised. Moreover, ATP is consumed by compensatory mechanisms attempting to restore NAD+ levels (29). Thus PARP hyperactivation, which is the central process in the cell death program called parthanatos (121) (discussed in sect. IIIC4), may also contribute to necrotic cell death.
Another potential amplification loop in necrosis is mediated by AIF, which functions as a mitochondrial oxidoreductase in healthy cells (77). We have previously noted that this protein has been inadvertently misnamed because its main cell death roles appear to be in necrosis and parthanatos rather than apoptosis (248, 334). PAR polymers from PARP-1 hyperactivation, calpains, and BAX can promote the release of AIF to the cytosol (334, 462, 552, 553). Following this, AIF translocates to the nucleus and promotes large-scale DNA cleavage (200 kb to 50 kb) as opposed to internucleosomal DNA cleavage mediated by CAD/DFF40 in apoptosis (115, 285, 287, 412). This DNA cleavage, which is carried out in partnership with macrophage migration inhibitory factor (MIF) (511), further activates PARP-1 resulting in more NAD+ consumption and energetic deficits in a positive feed-forward manner.
Interconnections exist between apoptosis and necrosis signaling. We have already considered how the differential usage of complexes IIa and IIb adjudicate the decision between apoptosis and necroptosis in the death receptor pathway. Although less defined, it is highly likely that apoptosis and necrosis also interconnect in the mitochondrial pathway (10). As previously mentioned, mPTP opening may lead to release of cytochrome c and caspase activation at late time points (17). This most likely results from the aforementioned osmotic effects of mPTP opening which promote OMM rupture rather than BAX/BAK-dependent MOMP. The extent to which this caspase activation contributes to the amplification of necrotic cell death, however, remains undetermined. Connections in the other direction (apoptosis → necrosis) may also exist. For example, during apoptosis, NADH dehydrogenase (ubiquinone oxidoreductase core subunit S1) Fe-S protein 1 (NDUFS1) undergoes caspase-mediated cleavage resulting in oxidative stress, mitochondrial depolarization, and swelling (401), events that may reflect necrosis. This raises the possibility that other mitochondrial proteins may also be caspase substrates. Further investigation is needed to delineate bidirectional cross-talk between apoptotic and necrotic signaling within the mitochondrial pathway.
III. REGULATED CELL DEATH IN HEART DISEASE
A. Overview of Myocardial Infarction and Heart Failure
Regulated and unregulated forms of cell death have been implicated in the pathogenesis of multiple forms of heart disease ranging from MI without reperfusion, MI with reperfusion (I/R), heart failure of diverse etiologies, myocarditis, congenital heart disease, and others (95, 167, 240, 327, 356, 366, 419, 435, 527). MI and heart failure have been studied most intensively, and we will focus on these syndromes in light of their high clinical relevance and because the available information allows them to be discussed mechanistically (TABLE 3).
Table 3.
Effects of death pathway manipulations in cardiac disease models and humans
Pathways | Cell Type | Cardiac Disease Model | Outcomes | Reference Nos. |
---|---|---|---|---|
Death receptor pathway in MI | ||||
Fas –/– (lpr mutant) | Global | I/R (in vivo) | Reduced infarct size | 258 |
Tnfr1 –/– | Global | MI without reperfusion (in vivo) | No change in infarct size | 250 |
Tnfr2 –/– | Global | MI without reperfusion (in vivo) | No change in infarct size | 250 |
Tnfr1 –/–; Tnfr2 –/– | Global | MI without reperfusion (in vivo) | Increased infarct size | 250 |
Necrostatin-1 (WT mice) | I/R (in vivo) | Reduced infarct size | 448 | |
Necrostatin-1 (global ppif –/– mice) | I/R (in vivo) | No additional reduction in infarct size | 272 | |
Ripk3 –/– | Global | I/R (in vivo) | Reduced infarct size | 351, 563 |
miR-103/107 antagomir | I/R (in vivo) | Reduced infarct size and necrosis | 507 | |
miR-873 | I/R (in vivo) | Reduced infarct size and necrosis | 508 | |
NRF-siRNA | I/R (in vivo) | Reduced infarct size and necrosis | 508 | |
miR-223 tg overexpression | Cardiomyocyte | I/R (in vivo) | Reduced infarct size and necrosis | 388 |
miR-223 –/– | Global | I/R (in vivo) | Increased infarct size and necrosis | 388 |
Mitochondrial pathway in MI | ||||
Bcl-2 tg overexpression | Cardiomyocyte | I/R (in vivo) | Reduced infarct size | 38, 54 |
Bax –/– | Global | I/R (isolated perfused) | Reduced infarct size, apoptosis, and necrosis | 192 |
Bax –/– | Global | MI without reperfusion (in vivo) | Reduced infarct size and post MI dysfunction | 191 |
Bax –/–; Bak –/– | Bax (cardiomyocyte) | I/R (in vivo) | Reduced infarct size, apoptosis, and necrosis | 528 |
Bak (global) | ||||
Ppif –/– | Global | I/R (in vivo) | Reduced infarct size and necrosis | 17, 341 |
Ppif tg overexpression | Cardiomyocyte | Baseline (in vivo) | Necrosis | 17 |
Ppif –/– + Bax –/–; Bak –/– | Bax (cardiomyocyte) | I/R (in vivo) | No additional reduction in infarct size | 528 |
Bak (global) | ||||
Ppif (global) | ||||
Puma –/– | Global | I/R (isolated perfused) | Reduced infarct size, apoptosis, and necrosis | 483 |
Cyclosporine A | I/R (various models) | Reduced infarct size (with some variability) | 367, 273 | |
Cyclosporine A (humans) | I/R (in vivo) | Reduced infarct size | 381 | |
Cyclosporine A (humans) | I/R (in vivo) | No reduction in infarct size or post MI dysfunction | 69 | |
Death receptor and mitochondrial pathways in MI | ||||
cIAP2 tg overexpression | Cardiomyocyte | I/R (in vivo) | Reduced infarct size, apoptosis, and necrosis | 62 |
UCF-101 | I/R (in vivo) | Reduced infarct size | 24, 282 | |
ARC tg overexpression | Cardiomyocyte | I/R (in vivo) | Reduced infarct size | 386 |
Caspase inhibitors | I/R (in vivo) | Reduced infarct size | 195, 198, 538, 544 | |
Procaspase-3 –/– | Procaspase-3 (cardiomyocyte) | I/R (in vivo) | No reduction in infarct size or post MI dysfunction | 206 |
Procaspase-7 –/– | Procaspase-7 (global) | |||
Death receptor pathway in HF | ||||
Procaspase-8 tg overexpression | Cardiomyocyte | Baseline (in vivo) | 0.023% TUNEL+ cardiomyocytes (control 0.002%) | 524 |
(limited activation) | Lethal heart failure | |||
Procaspase-8 tg overexpression | Cardiomyocyte | Baseline (in vivo) | Rescue of TUNEL positivity and heart failure | 524 |
(limited activation + delayed caspase inhibition) | ||||
Traf2 –/– | Cardiomyocyte | Baseline (in vivo) | Dilated cardiomyopathy and heart failure | 169 |
Traf2 +/– | Cardiomyocyte | MI without reperfusion (in vivo) | Dilated cardiomyopathy and heart failure | 169 |
Traf2 –/–; Tnfr1 –/– | Traf2 (cardiomyocyte) | Baseline (in vivo) | Rescued pathological phenotype of Traf2 –/– | 169 |
Tnfr1 (global) | ||||
Traf2 –/–; Ripk3 –/– | Traf2 (cardiomyocyte) | Baseline (in vivo) | Rescued pathological phenotype of Traf2 –/– | 169 |
Ripk3 (global) | ||||
Tak1 –/– | Cardiomyocyte | Baseline (in vivo) | Dilated cardiomyopathy and heart failure | 265 |
Tak1 –/–; Tnfr1 –/– | Tak1 (cardiomyocyte) | Baseline (in vivo) | Rescued pathological phenotype of Tak1 –/– | 265 |
Tnfr1 (global) | ||||
Ripk3 –/– | Global | MI without reperfusion (in vivo) | Reduced cardiac remodeling and heart failure | 298 |
Ripk3 –/– | Global | Doxorubicin | Reduced cardiac dysfunction | 563 |
Mitochondrial pathway in HF | ||||
L-type Ca2+channel (LTCC) tg overexpression | Cardiomyocyte | Baseline (in vivo) | Ca2+ overload, necrosis, heart failure, premature death. | 346 |
LTCC tg overexpression | LTCC (cardiomyocyte) | Baseline (in vivo) | Rescued heart failure | 346 |
Ppif –/– | Ppif (global) | |||
Ppif –/– | Global | Doxorubicin | Decreased cardiac dysfunction and necrosis | 346 |
Ppif -/ | Global | Pressure overload | Dilated cardiomyopathy and heart failure | 114 |
Shift from fatty acid to glucose use | ||||
Ppif –/– | Global | Swimming | Mortality in 5 days | 114 |
Increased hypertrophy and pulmonary edema | ||||
Bnip3 –/– | Global | I/R (in vivo) | Reduced apoptosis, cardiac remodeling, and heart failure | 96 |
Bnip3 –/– | Global | Doxorubicin | Reduced mitochondrial abnormalities and mortality | 90 |
Bnip3 tg overexpression | Cardiomyocyte | Baseline (in vivo) | Pathological remodeling and heart failure | 96 |
Gαq tg overexpression | Cardiomyocyte | Baseline (in vivo) | Increased apoptosis, hypertrophy, and heart failure | 3, 71 |
Gαq tg overexpression | Cardiomyocyte | Pregnancy | Heart failure | 3 |
Gαq tg overexpression + caspase inhibition | Cardiomyocyte | Pregnancy | Rescued heart failure | 180 |
NIX/BNIP3L tg overexpression | Cardiomyocyte | Baseline (in vivo) | Increased apoptosis and heart failure | 557 |
Gαq tg overexpression | Cardiomyocyte | Baseline (in vivo) | Rescued apoptosis and heart failure | 557 |
Autophagy-dependent cell death | ||||
Beclin-1 +/– | Global | Pressure overload | Reduced autophagy and cardiac dysfunction | 570 |
Beclin-1 tg overexpression | Cardiomyocyte | Pressure overload | Increased autophagy and cardiac dysfunction | 570 |
Beclin-1 +/– | Global | I/R (in vivo) | Reduced autophagy and infarct size | 310 |
Beclin-1 +/– | Beclin (global) | Baseline (in vivo) | Accelerated heart failure and early mortality | 475 |
Desmin tg overexpression | Desmin (cardiomyocyte) | |||
Atg5 –/– (postnatal) | Cardiomyocyte | Baseline (in vivo) | Heart failure and TUNEL+ cardiomyocytes | 344 |
Atg5 –/– (embryonic) | Cardiomyocyte | Pressure overload | Heart failure | 344 |
Ferroptosis | ||||
Deferoxamine (iron chelator) | Perfusion | I/R (isolated perfused) | Reduced infarct size | 144 |
Glutaminolysis inhibitor | Perfusion | I/R (isolated perfused) | Reduced infarct size | 144 |
GPX4 tg overexpression (mitochondrial targeted) | Global | I/R (isolated perfused) | Reduced release of creatine kinase | 72 |
Ferrostatin-1 | I/R (in vivo) | Reduced infarct size and myocardial enzyme release | 120, 268 | |
Cardiac dysfunction and pathological remodeling | ||||
Ferrostatin-1 | Doxorubicin | Reduced cardiac dysfunction | 120 | |
Ferrostatin-1 | Heart transplantation | Reduced cardiac cell death | 268 | |
Dexrazoxane | I/R (in vivo) | Reduced infarct size and myocardial enzyme release | 120 | |
Dexrazoxane | Doxorubicin | Reduced cardiac dysfunction | 120 | |
Pyroptosis | ||||
Caspase-1 –/– | Global | I/R (in vivo) | Reduced infarct size | 229 |
Caspase-1 –/– | Global | MI without reperfusion (in vivo) | Reduced cardiac dysfunction | 317 |
Caspase-1 tg overexpression | Cardiomyocyte | Baseline (in vivo) | Heart failure | 317 |
Caspase-1 tg overexpression | Cardiomyocyte | I/R (in vivo) | Increased infarct size | 467 |
ASC –/– | Global | I/R (in vivo) | Reduced infarct size | 229 |
ASC –/– bone marrow | ASC (global) | I/R (in vivo) | Reduced infarct size compared with WT | 229 |
Into WT mice | Same infarct size compared with ASC –/– | |||
WT bone marrow | ASC (global) | I/R (in vivo) | Reduced infarct size compared with WT | 229 |
into ASC –/– mice | Same infarct size compared with ASC –/– | |||
Parthanatos | ||||
Parp-1 –/– | Global | I/R (isolated perfused) | Reduced cardiac contractile dysfunction | 165, 378, 567 |
Parp-1 –/– | Global | I/R (in vivo) | Reduced infarct size and myocardial enzyme release | 543, 573 |
PARP-1 inhibitor | I/R (in vivo) | Reduced infarct size | 269, 378, 517, 572 | |
PARP-1 inhibitor | Perfusion | I/R (isolated perfused) | Reduced cardiac contractile dysfunction | 98, 567 |
Parp-1 –/– | Global | Pressure overload | Reduced pathological remodeling | 379 |
Mif –/– | Global | I/R (in vivo) | Increased infarct size | 324, 387 |
Mif –/– | Global | I/R (isolated perfused) | Increased infarct size | 387 |
I/R, ischemia/reperfusion; MI, myocardial infarction; tg, transgenic.
1. Myocardial infarction
MI results from acute and prolonged deficits in the supply of oxygen, nutrients, and survival factors to the myocardium relative to the demands of this tissue. Although this imbalance can be created through various means, it is most commonly caused by sudden and prolonged ischemia. The latter often results from thrombotic occlusion of a coronary artery following rupture/erosion of an atherosclerotic plaque (270, 271). Total occlusion of a coronary artery manifests clinically as ST-segment elevation MI (STEMI), which is usually transmural at the pathological level. Infarction is accompanied by multiple structural and functional consequences, the most irreversible of which is myocardial cell death. This involves both cardiomyocytes and non-myocytes, although the latter is less well documented (423, 472, 473). Moreover, in contrast to other cardiac syndromes (e.g., heart failure), where multiple processes contribute to pathogenesis, cell death is the initiating and central cardiac event in MI (TABLE 3).
Work in the 1980s and 1990s established reperfusion therapy (initially shown with thrombolytic agents; later using angioplasty/stenting) as beneficial in reducing infarct size (340). Infarct size is critical because it is a major determinant of subsequent cardiac dysfunction/heart failure and mortality. Although the benefit of reperfusion in preventing these serious outcomes of MI is unquestionable, it is clear that the reperfusion process itself begets myocardial cell death through a combination of oxidative stress, Ca2+ overload, and inflammation (collectively referred to as reperfusion injury) (178, 549). While it is extremely difficult to estimate the relative magnitudes of cardiac damage resulting from reperfusion per se as opposed to the prolonged ischemia that precedes it, some investigators have proposed that reperfusion accounts for up to ~50% of the total myocardial damage (549). In animal models, reperfusion generally limits the transmural extent of infarction to the subendocardial/mid-myocardial portion of the area at risk.
Non-ST-segment MI (non-STEMI) results from acute obstruction of a coronary artery that is often subtotal or “flickering” between subtotal and total. This dynamicity results in some degree of reperfusion and the resulting pathological injury is classically subendocardial. Interestingly, the incidence of STEMI is decreasing, while that of non-STEMI has remained relatively constant (546), the causes of this shift being unclear but possibly related to healthier lifestyles, reduction of cardiac risk factors, or use of statins. Most MI studies in animals, however, involve coronary artery ligation without or with subsequent reperfusion and, thus, model non-reperfused or reperfused STEMI rather than non-STEMI.
In this section, we review genetic and pharmacological manipulations of various cell death pathways and their effects on infarct size and post-MI cardiac function. Some general points will be noted here. First, in most cases, genetic and pharmacological inhibition of cell death signaling reduces cardiomyocyte death and infarct size only in the context of reperfused MI (I/R). Such reductions are not usually seen in permanent ischemia models and, when reductions are observed in those contexts, the possibility of inadvertent reperfusion needs to be considered. Why are reductions in cardiomyocyte death and infarct size not achieved without reperfusion? Several possible explanations can be envisioned: 1) initial regulated cell death transitions to unregulated cell death if the death stimulus persists. 2) Cellular repair processes are initiated with reperfusion so that, if the initial ischemic killing can be reduced, the net effect is tissue salvage. 3) Inhibition of cell death signaling is actually mitigating reperfusion injury rather than ischemic damage. While suggestive information support each of these possibilities, definitive data are lacking. Moreover, these possibilities are not mutually exclusive.
Second, despite extensive research, it remains unclear which cell death programs are most important during MI. Studies predating the regulated cell death era have traditionally considered unregulated necrosis to be the primary mode of cardiomyocyte death in this syndrome based primarily on morphological criteria (211, 212). When apoptosis became recognized as the first regulated cell death process, multiple studies demonstrated that genetic manipulations that inhibit apoptosis signaling in the death receptor and mitochondrial pathways resulted in reductions in apoptosis markers (e.g., TUNEL) in cardiomyocytes and in infarct size in mice subjected to myocardial I/R (38, 54, 137, 156, 213, 258, 309, 320). These experiments were interpreted as showing that cardiomyocyte apoptosis is critical in the pathogenesis of MI with reperfusion. More recent studies have raised doubts about the relevance of apoptosis in cardiomyocyte loss during MI (206). Moreover, genetic inhibition of pathways that mediate mitochondrial-dependent necrosis (17, 341), necroptosis (258, 351, 563), ferroptosis (15, 72, 120, 144), pyroptosis (229), parthanatos (543), and autophagy-dependent cell death (290, 310) have also demonstrated reductions in cardiomyocytes death and infarct size in the context of I/R (TABLE 3). This raises the question as to why multiple death pathways are being activated during reperfused MI. Possible explanations include 1) multiple cell death processes are, in fact, operating during MI with some cells dying through one modality and other cells through another. 2) Overlap/connections among mechanisms and markers have confounded analyses that assign cell death to one or another program. 3) Redundancy in signaling mechanisms has resulted in another cell death program taking over when a given program is inhibited. These issues need to be sorted out possibly through combined manipulations of cell death pathways coupled with a more comprehensive analysis of pathway-specific markers.
2. Heart failure
Heart failure, which is a highly heterogeneous syndrome in terms of etiologies, is generally divided at the pathophysiological level into heart failure with reduced ejection fraction (HFrEF) and heart failure with preserved ejection fraction (HFpEF) (34). HFrEF is characterized by defects in cardiac contraction, whereas HFpEF is defined at the cardiac level primarily by impaired filling. The role of cardiomyocyte death has been explored almost exclusively in HFrEF, where the attrition of cardiomyocytes (referred to as “dropout”) has been noted.
Measurement of the magnitude of decreases in numbers of cardiomyocytes is highly controversial and fraught with methodological issues, which will not be reviewed here. Because of these limitations in quantifying numbers of cardiomyocytes, markers of cell death programs have been employed. The rate of TUNEL-positive cardiomyocytes in failing human hearts has been estimated to be 0.08–0.25% as compared with 0.001–0.002% in nonfailing hearts (167, 366, 418–420). While these numbers reflect only a single cell death marker, they are instructive in two respects. First, they show that rates of cardiomyocytes death are higher in failing versus nonfailing human hearts. Second, they show that the overall magnitude of cardiomyocyte death is quite low, raising the question as to whether cardiomyocyte death is important in the pathogenesis of heart failure. As will be discussed below, genetic manipulations of cell death pathways demonstrate that cardiomyocyte death is an important component in the progression to heart failure (TABLE 3). However, in contrast to MI where cardiomyocyte death is the central event, it is one component process among many in the pathogenesis of heart failure.
B. Apoptosis and Necrosis
1. Death receptor pathway in MI
A number of studies have examined the role of death receptor signaling in I/R (TABLE 3). Some pre-dated and some post-dated the recognition that this pathway stimulates necroptosis as well as apoptosis. Accordingly, the cell death markers analyzed and accompanying mechanistic experiments often do not definitively differentiate between these death programs.
The death ligand FasL is upregulated in the rodent myocardium during global I/R in isolated, perfused hearts, although the cell type of origin was not identified (213). To test the role of Fas signaling in the pathogenesis of I/R, mice deficient in Fas (lpr mutant with global absence of Fas) were studied at an age before they manifest baseline immunological abnormalities. Reductions in infarct size accompanied by decreases in the percentage of TUNEL-positive cardiomyocytes were observed following I/R in vivo (258). Reductions in TUNEL were interpreted as indicating modulation of apoptosis. Necroptosis readouts were not assessed. These data suggest that Fas signaling contributes to myocardial damage during I/R.
Similar experiments were performed using global TNFR knockout mice in non-reperfused MI. Ablation of either TNFR1 or TNFR2 did not alter infarct size compared with controls 24 h following permanent coronary occlusion (250). In contrast, combined deletion of both receptors increased infarct size. While it is possible that this result reflects loss of the pro-survival effects of TNFR1 at complex I, it is curious that simultaneous deletion of TNFR2 was needed to reveal this effect. TNFR2, which lacks a DD, is not thought to directly signal through complex I, although it may influence TRAFs, IAPs, and NF-κB through other mechanisms (122). In addition, this receptor is mainly present in immune cells where it activates cell proliferation (514), although there is some evidence that its expression is upregulated in the heart at least late after infarction (328). This study suggests that the overall effect of TNF-α signaling in non-reperfused MI is to limit infarct size, and either TNFR1 or TNFR2 suffices for this effect.
The small molecule inhibitor necrostatin-1 (and its optimized analog necrostatin-1s) allosterically inhibits RIPK1 kinase activity (81, 82) to block necroptosis. Necrostatin-1, given at the time of reperfusion, reduced infarct size in mice subjected to I/R in vivo (448). These findings were extended to an in vivo pig model that showed that necrostatin-1, administered 10 min before reperfusion, reduced infarct size and ameliorated cardiac dysfunction in a dose-dependent manner (242). Interestingly, necrostatin-1 did not provide further reduction in infarct size in ppif −/− mice (lacking cyclophilin D), which were already partially cardioprotected (272) as previously shown (17). These data raise the possibility that RIPK1 and cyclophilin D, a key regulator of mPTP, signal in the same pathway. While there is no biochemical evidence linking RIPK1 and cyclophilin D/mPTP, a connection has been found between RIPK3 (which can be activated by phosphorylated RIPK1) and mPTP-mediated necrosis (563), which will be discussed below. Another caveat that may be relevant to this study is that necrostain-1 has been reported to have off-target effects (26, 60, 134, 470), one of which involves inhibition of ferroptosis (134), a death program that also mediates I/R (72, 120, 144). Markers of ferroptosis were not assessed in this study. Nonetheless, these data suggest that necroptosis may mediate I/R.
Global deletion of RIPK3 limited infarct size following I/R (351, 563). Interestingly, Ca2+/calmodulin-dependent protein kinase IIδ (CaMKIIδ), a facilitator of mPTP-mediated necrosis (216, 571), was identified as a novel target of RIPK3 (563). RIPK3 activates CaMKIIδ through phosphorylation and indirectly through oxidation. Furthermore, pharmacological inhibition of CaMKII, a known meditator of mPTP opening and I/R injury, blocks mPTP opening and cardiomyocyte killing specifically in response to RIPK3 overexpression (563). While mitochondria do not appear essential for TNF-α-induced necroptosis in some systems (468), it remains possible that mPTP opening contributes to RIPK3-induced cardiomyocyte killing. Whether the RIPK3-CaMKIIδ connection obviates the need for MLKL in bringing about the necrosis phenotype was also explored with MLKL knockdown, but was inconclusive because the resulting depletion of MLKL was incomplete (563). These data suggest, however, that RIPK3 provides a connection between necroptosis and mPTP-mediated necrosis.
Several studies have tested the involvement of microRNAs (miRs) and long noncoding RNAs (lncRNAs) in regulating death receptor signaling during I/R. For example, miR-103/107 can be upregulated by oxidative stress in cell systems to decrease FADD abundance and unleash necroptosis (507). Expression of miR-103/107 is upregulated in the ischemic region during I/R in vivo, and administration of specific anti-miRs significantly attenuates necrosis, infarction, and improves cardiac function (507). miR-103/107 is functionally inhibited by the lncRNA H19. This may serve to mitigate the FADD lowering and necroptosis-promoting effects of miR-103/107, although the involvement of this lncRNA was not tested in vivo. In contrast to miR-103/107, which promotes necroptosis, miR-873 inhibits this cell death program by decreasing the abundance of RIPK1 and RIPK3 in cardiomyocytes (508). miR-873 is downregulated during I/R, and intracoronary delivery of miR-873 limits I/R-induced necrosis and infarct size. miR-873 abundance is regulated by a lncRNA named necrosis-related factor (NRF) that functions as a sponge. NRF abundance is upregulated during I/R, which is functionally important as its silencing decreases infarct size. Interestingly, NRF appears to be a transcriptional target of p53 (508). A third study identified miR-223 as a potential regulator of multiple components of the death receptor pathway, including TNFR1, Death Receptor 6 (DR6), and IKKα, as well as NOD-like receptor family pyrin domain containing 3 (NLRP3), an inflammasome component involved in pyroptosis (388). miR-223 abundance increased in the heart in response to I/R. Moreover, cardiac-specific transgenic overexpression of miR-223 reduced markers of necrosis (but not apoptosis) and infarct size, while global knockout did the opposite.
2. Mitochondrial pathway in MI
A number of studies have examined the effects of various BCL-2 proteins on I/R injury, including the pro-survival subfamily, the multidomain pro-cell death subfamily, and the BH3-only proteins. Again, many of these studies were performed before the realization that BAX and likely BAK can mediate necrosis, in addition to apoptosis, in the mitochondrial pathway (TABLE 3).
Two groups independently studied the effect of cardiac-restricted transgenic BCL-2 overexpression on I/R injury in vivo and found reductions in apoptotic markers and infarct size accompanied by amelioration of cardiac dysfunction (38, 54).
Despite the redundancy between BAX and BAK signaling in some cell systems (e.g., MEFs; Ref. 520), global deletion of BAX alone reduced infarct size, apoptotic and necrotic markers, and cardiac dysfunction following I/R in isolated, perfused hearts (192). The same group found that BAX deletion resulted in modest reductions in infarct size in non-reperfused MI in vivo accompanied by amelioration of cardiac dysfunction 28 days later (191). In a separate cohort of mice also subjected to non-reperfused MI, cardiac caspase-3 activity at 2 h (apoptosis marker) and release of cardiac enzymes into the blood at 24 h (necrosis markers) were also reduced by Bax deletion. These data show that the absence of BAX reduces infarct size and cardiac remodeling during MI.
As discussed previously, more recent work has demonstrated that, in addition to their traditional roles in promoting MOMP and apoptosis, BAX and perhaps BAK mediate primary necrosis in the mitochondrial pathway by sensitizing cells to Ca2+-induced mPTP opening (225, 528). Mice with cardiomyocyte-restricted deletion of Bax and global deletion of Bak exhibited reduced infarct size following I/R compared with wild-type mice, accompanied by reductions in morphological features of necrosis, including mitochondrial abnormalities and derangement of sarcomere structure, and infarct size (528). Additionally, global deletion of ppif (encoding cyclophilin D) in the context of the cardiomyocyte-restricted Bax and global Bak knockout (triple knockouts) did not result in additional reduction in infarct size (528). These data provide genetic evidence that BAX/BAK signaling resides in the same (or an overlapping) pathway with cyclophilin D, a positive regulator of mPTP opening. This result further supports the notion that BAX/BAK signal necrosis in this context.
PUMA, which is transcriptionally induced by p53, is a BH3-only protein that functions as a direct activator of BAX and BAK (57, 397). Global deletion of PUMA reduced infarct size in isolated, perfused hearts subjected to I/R, which was accompanied by decreases in apoptosis and necrosis markers and lessening of cardiac dysfunction (483).
The defining feature of mitochondrial-mediated necrosis, Ca2+-induced opening of the mPTP, is facilitated by cyclophilin D. Two groups independently created global knockout mice lacking cyclophilin D and observed inhibition of Ca2+-induced mPTP opening and necrotic cell death (17, 341). Apoptotic cell death was unaffected. Moreover, absence of cyclophilin D markedly reduced infarct size in response to I/R. Conversely, cardiac-specific overexpression of cyclophilin D caused mitochondrial swelling and cell death (17). These studies provided the initial genetic evidence that necrosis mediated through the mitochondrial pathway is important in MI.
The cyclophilin D inhibitor CsA reduces Ca2+-induced mPTP opening and anoxic injury in cardiomyocytes (350). Multiple studies have shown that CsA, and analogs, ameliorates myocardial damage during I/R in various animal models (367), albeit with some variability in results (273). This drug was also suggested to reduce infarct size during reperfused MI in humans, although the study involved a small number of subjects (381). Ultimately, however, the most definitive human study to date failed to show reductions in cardiac remodeling, heart failure, and mortality, the most clinically relevant end points (69). This lack of efficacy probably reflects the fact that reduction of infarct size was not replicated in this cohort (69, 278). As will be discussed below, these data do not negate the possibility that cyclophilin D or mPTP is an important therapeutic target in I/R. Rather, they raise the possibilities that CsA may not be the optimal drug and/or that other death pathways need to be simultaneously blocked.
3. Molecules that operate in both death receptor and mitochondrial pathways in MI
As previously discussed, cIAP1, cIAP2, and XIAP play roles in both the death receptor and mitochondrial pathways. In the death receptor pathway, they activate survival pathways through ubiquitination events in complex I that suppress apoptosis and necroptosis (434). In the mitochondrial pathway, IAPs limit apoptosis by inhibiting caspases (87, 89, 108, 407). cIAP2 is upregulated by ischemia in the heart, and transgenic mice with cardiac-specific overexpression of cIAP2 exhibited modest reductions in infarct size following I/R (62) (TABLE 3). This was accompanied by reductions in both blood levels of cardiac enzymes (a marker of necrosis) and fewer TUNEL-positive cells (apoptosis). The study did not explore the relative extents to which these effects were mediated through inhibition of death receptor or mitochondrial pathways.
As previously discussed, the mitochondrial apoptogens SMAC/DIABLO and OMI/HtrA2 relieve IAP-mediated inhibition of caspases by directly binding IAPs and displacing caspases (185, 453, 464). In addition, OMI/HtrA2 also cleaves and permanently inactivates XIAP through its serine protease function (118, 464, 537). UCF-101, a small molecule inhibitor of the OMI/HtrA2 serine protease activity, was shown to reduce XIAP loss, caspase activation, and infarct size following I/R when administered 10 min before reperfusion (24, 282). The efficacy of UCF-101 in limiting infarct size is notable because its presumed site of action is postmitochondrial, meaning after potential mitochondrial dysfunction has been induced during apoptosis. This raises the possibility that survival may be promoted through additional mechanisms such as IAP-mediated effects at complex I.
Apoptosis repressor with caspase recruitment domain (ARC) (149, 241), a CARD-containing protein that is highly expressed in cardiomyocytes, is a potent inhibitor of several cell death programs (131, 313, 347). ARC blocks death receptor-mediated apoptosis through interactions with FADD that interfere with DISC formation (347). ARC suppresses mitochondrial-mediated apoptosis by inhibiting BAX conformational activation through direct binding (347). An additional mechanism by which ARC inhibits mitochondrial death signaling is through its interaction with p53 in the nucleus (131). This inhibits p53 tetramerization, which both interferes with p53 function as a transcription factor and promotes export from the nucleus. Finally, ARC attenuates the ER stress response in some cell types through mechanisms involving CHOP (313). Although ARC undergoes proteasomal degradation during oxidative stress and I/R (130, 348), cardiomyocyte-specific transgenic overexpression reduces infarct size (386). Global ARC deletion has also been reported to exacerbate infarct size following I/R (99). But this study was performed in a small number of mice of mixed genetic background and has not been replicated using large numbers of genetically homogeneous mice (R. N. Kitsis, unpublished data). The lack of an effect of ARC deletion on increasing infarct size may reflect that endogenous ARC is also degraded during I/R in wild-type mice.
Various caspase inhibitors (typically poly- or pan-caspase inhibitors) have been shown to variably reduce infarct size in mice subjected to I/R accompanied by amelioration of cardiac dysfunction (195, 198, 538, 544). Although decreases in apoptosis markers were observed in most studies, the precise caspases that were antagonized and death programs that were blocked were not defined in most cases. Of particular interest is whether caspase inhibition attenuated mitochondrial dysfunction during I/R and, if so, what is the mechanism. Additionally, among the myriad substrates that are cleaved by caspase-3 during apoptosis, some are contractile proteins such as troponin T and α-actinin (65), the preservation of which by caspase inhibition could potentially lessen cardiac dysfunction.
While the above experiments showed some efficacy of caspase inhibitors to decrease infarct size following I/R, one study using combined deletion of procaspases-3 and -7 failed to show any effect on acute infarct size, postinfarct heart failure, or cardiomyocyte death, also using an I/R model (206). This study is important because caspases-3 and -7 are the major effector caspases. A limitation of this study, however, is that the third effector caspase, caspase-6, was not eliminated. Although its repertoire of substrates is more limited than that of caspases-3 and -7, it is difficult to know whether some compensation occurred.
4. Death receptor pathway in heart failure
As previously noted, 0.08–0.25% of cardiomyocytes in failing human hearts are TUNEL-positive compared with 0.001–0.002% in controls (167, 366, 418–420). A gain-of-function approach was used to assess the significance of low, but abnormal, rates of cardiomyocyte death in the pathogenesis of heart failure. Transgenic mice were created with cardiomyocyte-specific overexpression of an inducible procaspase-8 mutant (524) (TABLE 3). This mutant encoded a fusion protein consisting of a myristoylation signal (for plasma membrane targeting), three FK506 binding protein modules (FKBP), and the p20 and p10 subunits of procaspase-8. It had previously been shown in cell culture that addition of a small molecule that binds the FKBP modules could activate procaspase-8 by forced proximity (339). When this small molecule was administered to mice, they died acutely due to massive cardiomyocyte death, a result that is not informative about the pathogenesis of human heart failure. However, when left untreated for several months, spontaneous heart failure and mortality were noted in these transgenic mice, but not in wild-type mice or mice expressing a catalytically inactive version of the same transgene. Importantly, rates of TUNEL-positive cardiomyocytes in the mice dying of heart failure were 0.023% compared with 0.002% in wild-type mice and those with the catalytically dead mutant. Moreover, delayed administration of a pan-caspase inhibitor to the transgenic mice destined for heart failure rescued both cell death and heart failure. This study demonstrated that rates of cardiomyocyte death that are even 4- to 10-fold lower than those observed in failing human hearts are adequate to cause a lethal dilated cardiomyopathy. It should be noted that this study used caspase-8 activation as an experimental tool to address this question, but did not explore the role of caspase-8 and the death receptor pathway per se in mediating heart failure in response to various pathological stimuli.
TRAF2 is usually thought to be an adaptor protein in complex I, but it also contains a RING domain with E3-ligase activity (7, 259). While cIAP1/2 and LUBAC are generally considered the major enzymes in complex I responsible for adding K63-linked and linear ubiquitin chains, respectively, to RIPK1, it is possible that TRAF2 also plays a role. Cardiomyocyte-specific transgenic expression of TRAF2 activates NF-κB (41). Furthermore, cardiomyocyte-specific deletion of TRAF2 in mice, or expression of a TRAF2 mutant lacking the RING domain (which appears to function as a dominant negative) in neonatal cardiomyocytes, induces apoptosis and necrosis (169). The apoptosis component is sensitive to pharmacological inhibition of RIPK1 kinase activity by necrostatin-1. The necrosis component is mitigated by RIPK3 absence or MLKL knockdown. A constitutively active TAK1 mutant protects against both apoptosis and necrosis resulting from the TRAF2 dominant negative mutant expression, similar to the baseline suppression of both of these forms of cell death in vivo by endogenous TAK1 (265). While the effects of the dominant negative TRAF2 mutant suggest that its E3-ligase function is important in suppressing cell death at least in the heart, the precise mechanism remains to be defined. Potentially relevant TRAF2 substrates include RIPK1 and TAK1 (119, 562), but their roles remain to be tested. The preceding mechanism may be important in the pathogenesis of heart failure because TRAF2 abundance increases in response to pressure overload and post-MI heart failure, and cardiomyocyte-specific TRAF2 deletion induces dilated cardiomyopathy and heart failure (169). Thus TRAF2 appears important in the heart in pathological, as well as baseline, conditions.
The role of RIPK3 in heart failure was tested using a non-reperfused MI model in which infarct size was unchanged (298). Global RIPK3 knockout reduced adverse cardiac remodeling and dysfunction at 30 days. Markers of necroptosis signaling were not assessed in this study. The anthracycline doxorubicin, a chemotherapeutic agent used to treat multiple solid tumors and leukemias in adults and children, causes a dose-dependent cardiomyopathy in ~10% of patients. While the etiology of doxorubicin-induced cardiomyopathy is complex, apoptotic and necrotic cell death is believed to be involved. Global absence of RIPK3 also attenuated doxorubicin-induced cardiac dysfunction (563).
5. Mitochondrial pathway in heart failure
In light of the previously discussed role of cyclophilin D in mPTP opening and necrosis during I/R (341), the role of this pathway was also tested in heart failure models. Given the importance of Ca2+ as a trigger for mPTP opening, this was first done in mice with cardiac Ca2+ overload. This was induced by cardiomyocyte-specific overexpression of the β2a subunit of the plasma membrane L-type Ca2+ channel (LTCC) (346) (TABLE 3). These mice exhibited cardiomyocyte Ca2+ overload, cardiomyocyte necrosis, heart failure, and premature death. To test the importance of necrosis in causing heart failure, the transgenic mice overexpressing the β2a subunit of the LTCC were crossed with global ppif −/− mice. Interestingly, heart failure was rescued by the absence of cyclophilin D, but not by overexpression of BCL-2, implicating necrotic cell death in pathogenesis. In the same study, cyclophilin D loss was also shown to rescue fibrosis (as a surrogate for necrosis) and cardiac dysfunction resulting from administration of doxorubicin (346). This experiment suggests that the role of cyclophilin D and necrotic cell death in heart failure may extend beyond just Ca2+ overload and implicates mitochondrial-mediated cardiomyocyte necrosis in heart failure.
To further extend the role of cyclophilin D/mPTP and cardiomyocyte necrosis to other clinically relevant heart failure models, ppif −/− mice were subjected to pressure overload, a stress common to heart failure of multiple etiologies (114). Unexpectedly, pressure overload in ppif −/− mice resulted in marked systolic dysfunction accompanied by chamber dilation within 7 days, while wild-type controls manifest only relatively mild cardiac dysfunction at 6 wk. Pressure overload did not significantly increase the frequency of TUNEL-positive cardiomyocytes in either ppif −/− or wild-type mice. Thus the heart failure phenotype exhibited by ppif −/− mice was not cell death related. In contrast, transcript profiling in the baseline state suggested changes in substrate utilization away from fatty acids and toward glucose in ppif −/− mice, a metabolic change often observed in failing hearts (127). This effect was hypothesized to result from impaired Ca2+ efflux from the mitochondrial matrix in ppif −/− mice resulting in activation of Ca2+-dependent dehydrogenases. Interestingly, a swimming regimen, a stimulus that induces physiological cardiac hypertrophy with maintained or augmented systolic function, induced mortality in ppif −/− mice within 5 days. An exaggerated hypertrophic response accompanied by pulmonary edema was observed in ppif −/−, but not wild-type, mice. This study demonstrates that complex metabolic changes leading to heart failure are induced by pathological and physiological hypertrophic stimuli in the absence of cyclophilin D.
BCL-2/adenovirus E1B 19-kDa interacting protein 3 (BNIP3) and 19-kDa interacting protein 3-like protein X/BCL-2/adenovirus E1B 19-kDa interacting protein 3-like (NIX/BNIP3L) are two related BH3-only-like proteins that mediate a complex mixture of apoptotic and necrotic cell death (36, 49, 311). Each of these proteins also serves as adaptors between ubiquitinated OMM proteins and microtubule-associated protein 1 light chain 3 (LC3) in some mitophagy paradigms (561). The precise mechanisms by which BNIP3 and NIX/BNIP3L induce cell death are not understood, nor is the relationship between the cell death and mitophagy functions. Unlike classic BH3-only proteins, the BH3 domain of BNIP3 and NIX/BNIP3L is not required for cell death. Rather, mitochondrial targeting, which is mediated by the transmembrane domain in each protein, is obligatory for this function. BNIP3 and NIX/BNIP3L are each expressed as two isoforms generated by alternative splicing. The long isoform contains the transmembrane domain and induces cell death. The short isoform lacks the transmembrane domain and heterodimerizes with the long isoform to antagonize cell death. The relevance of these proteins to cardiac disease resides in their regulation by pathological stimuli. BNIP3 abundance increases at the transcriptional level in response to hypoxia (168, 396), while NIX/BNIP3L is transcriptionally upregulated by signaling mediated by Gαq, a protein coupled to certain cell surface receptors (angiotensin II type 1 receptor, endothelin receptor, and α1-adrenergic receptor) that mediate cardiac hypertrophy (557).
BNIP3 appears to induce primarily apoptotic cardiomyocyte death during cardiac remodeling post-I/R (96) and necrotic cardiomyocyte death following doxorubicin treatment (90). With regard to the I/R studies (96), cardiomyocyte-specific deletion of Bnip3 does not influence infarct size, perhaps because it is not yet fully induced by hypoxia in the time frame of cardiomyocyte killing in this model. However, cardiomyocyte-restricted absence of this protein decreases post-I/R apoptotic cell death in the peri-infarct zone and remote myocardium, thereby attenuating adverse cardiac remodeling and heart failure. Conversely, inducible, cardiomyocyte-specific transgenic overexpression of BNIP3 stimulates pathological modeling and heart failure even in the absence of infarction. Doxorubicin also induces BNIP3 expression in cardiomyocytes, and deletion of Bnip3 in these cells in vivo protects against abnormalities in mitochondrial morphology and respiration and necrotic cell death induced by doxorubicin (90). One relevant mechanism appears to be that the absence of BNIP3 prevents doxorubicin-induced disruption of an interaction between cytochrome c oxidase subunit 1 (COX1) and uncoupling protein 3 (UCP3), two proteins that reside in the IMM.
NIX/BNIP3L is important in mediating cardiomyocyte apoptosis mediated through Gαq (557). Transgenic mice with cardiac-restricted overexpression of Gαq develop cardiac hypertrophy, cardiomyocyte apoptosis, and heart failure (3, 71). In addition, a proportion of mice go into heart failure during pregnancy (3), likely induced by the hemodynamic stresses of pregnancy, rather than the vascular pathology that operates in human peripartum cardiomyopathy (376). Pregnancy-related heart failure in Gαq transgenic mice can be rescued by caspase inhibition (180). As previously noted, NIX/BNIP3L is upregulated by Gαq. Cardiomyocyte-restricted transgenic overexpression of NIX/BNIP3L phenocopies the cardiomyocyte apoptosis and heart failure exhibited by Gαq transgenic mice (557). Moreover, transgenic overexpression of the short inhibitory isoform of NIX/BNIP3L attenuates cardiomyocyte apoptosis and heart failure in Gαq transgenic mice. A proportion of NIX/BNIP3L may reside at the ER/SR, as well as at the mitochondria. Selective targeting of NIX/BNIP3L to mitochondria appears to induce primarily BAX/BAK and caspase-dependent apoptotic death, while targeting to ER/SR seems to induce primarily mPTP-dependent necrosis (53).
C. Other Death Programs
1. Autophagy-dependent cell death
Autophagy is an evolutionarily conserved process that degrades cellular components in the lysosome. It plays important roles in maintaining quality control at baseline and is augmented in response to stress to restore cellular homeostasis (326). Given its role in attenuating cellular stress, autophagy generally functions as a survival process (261). However, some evidence suggests that, under certain circumstances, autophagy may beget cell death. Autophagy-dependent cell death remains a controversial concept. It is defined as a regulated form of cell death that is caused by autophagy, as opposed to occurring in parallel with, or as a compensation for, autophagy (142). It remains unclear if autophagy-dependent cell death results from excessive rates of autophagy (possibly reflecting excessive catabolism of cellular components) or whether it is attributable to a qualitative change in the nature of the autophagy process itself (FIGURE 5). It should be noted that autophagy-dependent cell death is not synonymous with another regulated cell death program termed lysosome-dependent cell death (142, 433), which is caused by the release of lysosomal enzymes into the cytosol resulting from lysosomal membrane permeabilization. In addition, it is not clear if leakiness of the lysosomal membrane is a feature of autophagy-dependent cell death.
FIGURE 5.
Autophagy-dependent cell death. The “conventional” view of autophagy-dependent cell death is that increases in rates of macroautophagy induce cell death through excessive catabolism of cellular components. Specific death machinery and specific autophagy-dependent cell death markers have not been identified. In contrast, autosis is a form of autophagy-dependent cell death that is characterized by a specific cellular morphology (see text) and mediated by the plasma membrane Na+-K+-ATPase.
Before considering autophagy-dependent cell death, it should be noted that several autophagy programs have been described including macroautophagy, chaperone-mediated autophagy, microautophagy, and endosomal microautophagy (139). Studies pertaining to autophagy-dependent cell death have focused on macroautophagy, which will be referred to hereafter simply as autophagy. This process degrades substrates including macromolecules (e.g., proteins, lipids, and nucleic acids) and organelles by transporting them to lysosomes in double membrane vesicles termed autophagosomes. Autophagosomes subsequently fuse with the lysosomal membrane delivering their cargo to the lysosomal lumen where acidic hydrolases reside. Detailed descriptions of this process can be found elsewhere (139, 326). The selection of substrates in autophagy may be nonselective (in bulk) or selective, an example of the latter being the mitophagic removal of damaged mitochondria, which are marked by ubiquitination of OMM proteins. Autophagy is activated in response to diverse cellular stresses including starvation, ischemia, oxidative stress, genotoxic stress, proteotoxic stress, and infection.
Autophagy-dependent cell death was historically defined as “evidence of cell death” accompanied by cytoplasmic autophagic vacuoles (e.g., autophagosomes), these features most easily identified by electron microscopy (80). To reemphasize, however, this morphological definition does not discriminate among the possibilities that cell death is 1) being caused by autophagy, 2) accompanying autophagy but unrelated, or 3) inducing autophagy as a compensatory mechanism. The deficiencies of this morphological definition underscore the importance of 1) establishing cause-and-effect between autophagy and cell death and 2) having specific death readouts that reflect autophagy-dependent cell death. As described below in the heart, the cause-and-effect issue has been addressed by the studies in which autophagy is perturbed genetically following which cell death is assessed. However, death readouts specific for autophagy-dependent cell death still have not been identified, with the exception of a subset of autophagy-dependent cell death termed autosis (290) that will be discussed below.
In the heart, autophagy can be activated by prolonged ischemia, I/R, and during heart failure of various etiologies (475, 570). To test whether autophagy is responsible for cell death and the pathogenesis of these syndromes, the general strategy has been to genetically modulate autophagy in these disease contexts (TABLE 3). Beclin 1 is critical for early autophagosome formation. Global deletion of a single Beclin 1 allele (as biallelic deletion in the germ line is lethal; Ref. 556) ameliorates cardiac dysfunction resulting from severe pressure overload (570). Conversely, cardiomyocyte-specific Beclin 1 overexpression does the opposite. The expected effects of these Beclin 1 manipulations on rates of autophagy were confirmed using various autophagy markers. The only cell death readout in these studies was TUNEL, a marker of apoptotic cell death. However, TUNEL was not informative in this particular context because, as is frequently observed, pressure overload did not significantly increase the frequency of TUNEL-positive cardiomyocytes even in wild-type mice. These data show that increases in Beclin 1, resulting in augmentation of rates of autophagy, exacerbate cardiac dysfunction in this severe pressure overload model. Although this effect may be occurring through increases in cell death, this was not shown explicitly. That said, this latter criticism is addressed to some extent in a second study that used the same global Beclin 1 heterozygote knockout mouse in an I/R model (310). Loss of one Beclin 1 allele reduced infarct size and frequency of TUNEL-positive cardiomyocytes, thus providing more direct evidence that autophagy may exacerbate cardiomyocyte death under some pathological conditions. It should be noted, however, that in the setting of ischemia without reperfusion, stimulation of autophagy appears to be protective (310, 427, 428).
To add further confusion, however, a third study inhibited autophagy under basal and pressure overload conditions using a different genetic approach and obtained the opposite result (344). Autophagy related 5 (Atg5), which is involved in autophagosome maturation, was deleted specifically in adult cardiomyocytes in vivo. This spontaneously resulted in severe systolic dysfunction and heart failure within 1 wk accompanied by increased frequency of TUNEL-positive cardiomyocytes. Cardiomyocyte-specific deletion of Atg5 in embryonic life caused no baseline phenotype, but severe heart failure developed within 1 wk of pressure overload. These data suggest that endogenous levels of autophagy are required for normal systolic function under basal and pressure overload conditions, and reductions in autophagy under these conditions are accompanied by increased cardiomyocyte death.
How can we reconcile these two opposing data? One obvious difference is that different genes were manipulated, with Beclin 1 loss of function ameliorating cardiac pathology (pressure overload and I/R) versus Atg5 loss of function causing pathology at baseline and with pressure overload. Arguing against this explanation, however, is a fourth study showing that heterozygous global deletion of Beclin 1 exacerbates desmin-related cardiomyopathy (475). Nevertheless, the notion that differences in Beclin 1 and Atg5 may underlie these discrepant results is not resolved. In addition to its distinct role in autophagosome formation, Beclin 1 is also a BH3-only protein that is bound and negatively regulated by BCL-2 (363, 377). While it is generally believed that the Beclin 1-BCL-2 interaction does not interfere with the anti-cell death properties of BCL-2, some data suggest that enhancement of Beclin 1-BCL-2 or Beclin 1-BCL-xL binding resulting from phosphorylation of the BH3 domain of Beclin 1 may activate BAX (300). This raises speculation as to whether loss of Beclin 1, but not Atg5, somehow mitigates cell death. A second difference between the Beclin 1 and Atg5 loss-of-function studies is that the Beclin 1 gene was deleted in a global manner and Atg5 in a cardiomyocyte-specific manner. Insufficient information exists to assess the significance of this difference. A third difference is that the severity of pressure overload may have been greater in the Beclin +/− versus Atg5 −/− study, again a consideration that cannot yet be resolved. In summary, we are unable to reconcile these discrepant results. Moreover, these studies connect changes in macroautophagy most convincingly with changes in cardiac function rather than in autophagy-dependent cell death, making it difficult to draw conclusions specifically about effects on cell death.
a) autosis.
Recently, a new program of autophagy-dependent cell death termed autosis was described (290). Autosis is characterized by morphological changes distinct from those of apoptosis or necrosis that include early nuclear convolution followed by rapid ballooning of the perinuclear space. Autosis induction tracks with increased rates of autophagy and is prevented by pharmacological or genetic inhibition of autophagy, but not by inhibition of apoptosis, necrosis, or even lysosomal enzymes. Autosis can be induced by nutrient deprivation in cell culture and by hypoxia/ischemia in the brain. Most interestingly, autosis appears to be mediated by the plasma membrane Na+-K+-ATPase, and neuroprotection results from pharmacological inhibition of this molecule (FIGURE 5). These are early days for autosis, and clearly more work will be needed to understand its mechanism and potential significance as a cell death process in various contexts including heart disease.
2. Ferroptosis
Ferroptosis is an iron-dependent form of cell death characterized by oxidative damage to cellular membranes (97, 142) (FIGURE 6). Fenton reaction-generated ferric iron (Fe3+) increases ROS and activates lipoxygenases that damage cellular membranes [especially phosphatidyl ethanolamine-containing polyunsaturated fatty acids (PUFAs)] (539). Lipid peroxidation can be prevented, and ferroptosis blocked, by molecules that either chelate iron (e.g., deferoxamine, dexrazoxane) or oppose lipid peroxidation (e.g., ferrostatin-1, liproxstatin-1, vitamin E) (97, 134). The major endogenous mechanism to combat lipid peroxidation is glutathione peroxidase 4 (GPX4) (51, 308). GPX4 function requires the cofactor glutathione (GSH) and is therefore dependent on the plasma membrane cystine/glutamate antiporter system xc− because cystine is needed to generate GSH (97). Molecules that inhibit system xc− (e.g., erastin, sulfasalazine, sorafenib, glutamate) deplete the cellular pool of GSH and inhibit GPX4 function leading to ferroptosis (540). The morphological manifestations of ferroptosis are distinct from those of apoptosis and necrosis and include dense, compact mitochondria with loss of cristae (97). Some studies have reported OMM rupture (134, 535), but this is not a consistent or characteristic feature (B. Stockwell, personal communication). Loss of plasma membrane integrity has been used as a readout for ferroptosis (1), but it is presently unclear whether this is part of the primary process or simply reflects late downstream effects of oxidative damage. In fact, one of the most important unanswered questions is how ferroptosis kills cells, specifically it is not clear as to which cellular membranes need to be damaged to bring about this form of cell death (123).
FIGURE 6.
Overview of ferroptosis. Ferroptosis is an iron-dependent form of regulated cell death mediated by lipid peroxidation of cellular membranes, although which damaged membranes induce cell death is not yet clear. Ferroptosis can be initiated by iron overload in which Fenton reaction-produced Fe3+ activates lipoxygenases or by mechanisms that inactivate glutathione peroxidase 4 (GPX4), which opposes lipid peroxidation. GPX4 can be inactivated by extracellular glutamate overload, which inhibits antiporter system xc− . System xc− normally imports cystine, which is converted to cysteine that subsequently generates glutathione (GSH), a cofactor for GPX4. Fe3+ imported through the transferrin receptor (TR) is converted to Fe2+ in endosomes by the metalloreductase six-transmembrane epithelial antigen of prostate 3 (Steap3) and released from endosomes by divalent metal transporter 1 (DMT1). Erastin, sulfasalazine, and sorafenib are small molecules that inhibit system xc− to induce ferroptosis. Ras synthetic lethal 3 (RSL3) and FIN56 are small molecules that inhibit GPX4 also inducing ferroptosis. Ferrostatin-1, liproxstatin-1, vitamin E, and CoQ10 are lipid antioxidants that inhibit ferroptosis. In addition, FIN56 also inhibits CoQ10 through the mevalonate pathway. Deferoxamine (DFO) binds Fe2+ mainly extracellularly, while dexrazoxane and ciclopirox (CPX) bind Fe2+ intracellularly.
Ferroptosis has been implicated in cell death in a variety of disease settings including treatment with cancer drugs (540), glutamate-induced neuronal cell death and neurodegenerative disease (97, 446), drug-induced liver damage (295), acute hypoxia-induced kidney injury (134, 279, 446), and I/R in the liver (134) and heart (72, 120, 144). A strong rationale for investigating ferroptotic death in the heart is the known cardiotoxicity of iron and the fact that reperfused MI in humans is associated with iron overload in the peri-infarct zone (40).
Five studies involving ferroptosis are directly relevant to the heart (15, 72, 120, 144) (TABLE 3). The first confirmed that iron overload takes place in cardiomyocytes and nonmyocytes during I/R in vivo (15). It then went on to demonstrate that ferroptosis can be induced in primary cultures of adult mouse cardiomyocytes using either Fe3+ or either of two small molecule activators of ferroptosis: the system xc− inhibitor erastin or the GPX4 inhibitor Ras synthetic lethal 3 (RSL3). The read-out employed for cell death was loss of plasma membrane integrity, which could be rescued by ferrostatin-1, a lipophilic antioxidant. Importantly, cell death occurred independently of caspase-3 activation or alterations in autophagy. While, as discussed above, loss of plasma membrane integrity is not specific for ferroptosis and may not even be part of the direct mechanism, the fact that cell death could be induced by erastin and RSL3, which activate specific steps in ferroptotic cell death, demonstrates that ferroptosis can be induced in cardiomyocytes. Since mammalian target of rapamycin (mTOR) has previously been shown to decrease infarct size in myocardial I/R in vivo (13) and because mTOR also impacts iron handling (166), this study also investigated the effects of mTOR overexpression and knockout on cell death induced by Fe3+ and ferroptosis agonists in cultured adult cardiomyocytes. mTOR overexpression reduced, and knockout increased, cell death induced by these stimuli, and the mechanism may involve mTOR modulation of ROS production.
A second study showed that the iron chelator deferoxamine reduced infarct size following global I/R in isolated, perfused mouse hearts (144). This same study, which implicated glutaminolysis in the pathogenesis of ferroptosis, showed that a small molecule inhibitor of glutaminases also reduced infarct size in the same system.
A third study, which was published before ferroptosis was discovered, provides complementary information (72). Again, using global I/R in isolated, perfused hearts, the data show that global transgenic overexpression of a mitochondrial-targeted mutant of GPX4 is cardioprotective as measured by reduced release of creatine kinase, an indicator of loss of cell membrane integrity. Mitochondrial-targeted GPX4 also reduced mitochondria lipid peroxidation, and lessened mitochondrial and contractile dysfunction post-I/R.
A fourth study demonstrated that treating mice with either ferrostatin-1, or the iron chelator dexrazoxane, reduced infarct size as well as serum markers of myocardial injury during I/R in vivo (120). These compounds also improved survival and prevented cardiac dysfunction in mice treated with doxorubicin, which induces mitochondrial iron load and lipid peroxidation.
A fifth study showed that ferrostatin-1 blocked cardiomyocyte death both in heart transplantation and the traditional coronary artery ligation I/R models in vivo (268). In the transplantation model, ferrostatin-1 reduced levels of hydroperoxy-arachidonoyl-phosphatidylethanolamine, a mediator of ferroptosis, and deaths of cardiomyocytes and cardiac fibroblasts, but not endothelial cells. Furthermore, ferroptosis was shown to promote neutrophil recruitment in the transplantation model through a TLR4/TRIF/type 1 interferon (IFN) signaling pathway specifically in coronary vascular endothelial cells. Since ferrostatin-1 inhibited neutrophil recruitment both in this model and the I/R model, inhibition of cardiomyocyte death may have occurred, in part, through antagonizing neutrophil-dependent mechanisms. However, ferrostatin-1 also reduced infarct size in isolated buffer-perfused hearts, indicating that a component of the rescue involved inhibition of ferroptosis in cardiac cells.
Taken together, these studies show that ferroptosis can take place in adult cardiomyocytes. In addition, they implicate ferroptosis in the pathogenesis of myocardial I/R. An important caveat, however, is that the conclusion that ferroptosis is responsible for cell death in this syndrome is based primarily on the observation that inhibition of myocardial damage results from molecules known to block ferroptosis. Data that would strengthen the involvement of ferroptosis in this paradigm would include markers of the specific molecular events that mediate ferroptosis. These might include increases in pathogenic modified fatty acids such as hydroperoxy-arachidonoyl-phosphatidylethanolamine (268) and inhibition of GPX4 or system xc− (97). The role of ferroptosis in other cardiac syndromes, such as heart failure, also merits exploration.
3. Pyroptosis
Pyroptosis is a regulated form of cell death that is closely tied to the innate immune response and characterized by permeabilization of the plasma membrane and extracellular release of inflammatory cytokines (FIGURE 7). Plasma membrane rupture during pyroptosis is mediated by gasdermin D (GSDMD) (184, 230, 439). In healthy cells, GSDMD is held functionally inactive through an intramolecular interaction between its COOH-terminal (inhibitory) and NH2-terminal (pro-death) domain. Proteolysis of GSDMD by inflammatory caspases (human caspases-1, -4, and -5 and mouse caspases-1 and -11) generates the functionally competent NH2-terminal cleavage product (GSDMD-NT) (94, 286). GSDMD-NT preferentially binds to components primarily found in the plasma membrane inner leaflet of mammalian cells (phosphatidylinositol phosphates and phosphatidylserine), and the inner and outer leaflets of bacterial cells (cardiolipin), but does not bind to components present in the outer leaflet of mammalian cells (phosphatidylethanolamine and phosphatidylcholine) (286). Through still largely undefined mechanisms, GSDMD-NT monomers oligomerize to form pores in the plasma membrane and cause the release of intracellular material inducing cell death (94, 286). Interestingly, GSDMD itself is also released from cells undergoing pyroptosis, but does not damage neighboring cells likely due to the aforementioned membrane component binding specificity that prohibits association with mammalian outer leaflets (286). Gasdermin E [GSDME; also known as Deafness, autosomal dominant 5 (DFNA5)] is a second member of the conserved gasdermin family that displays a similar membrane-perforating function resulting in pyroptotic-like cell death. Importantly, however, GSDME is processed into its active NH2-terminal form by caspase-3 rather than by inflammatory caspases as is the case for GSDMD (406, 512).
FIGURE 7.
Overview of pyroptosis. The sentinel event in pyroptosis is the creation of pores in the plasma membrane by gasdermin D (GSDMD). GSDMD is activated through cleavage by inflammatory caspases (caspases-1, -4, and -5 in humans). Procaspase-1 is activated in response to diverse stresses including infection, toxic insults impinging on the cell, and multiple intracellular stresses [e.g., reactive oxygen species (ROS), cytosolic DNA, and others]. Pathogen-associated molecular patterns (PAMPs) and danger-associated molecular patterns (DAMPs) resulting from these stresses promote assembly of inflammasomes, which are subcellular multiprotein complexes in which procaspase-1 activation takes place. This involves the sensing of PAMPs and DAMPs by pattern recognition receptors such as NOD-like receptor (NLR) pyrin domain (PYD) containing receptor 3 (NLRP3). NLRP3 subsequently recruits ASC, which recruits and activates procaspase-1 (see text for details). Other proteins besides NLRP3 can also function as the “apical” protein in inflammasome assembly. Inflammasome formation is “primed” through transcriptional and nontranscriptional mechanisms (see text for latter). Transcriptional priming occurs downstream of Toll-like receptors (TLRs) and TNFR1 through NF-κB-mediated increases in NLRP3, ASC, and procaspase-1 expression. In addition, the expression of downstream effectors in this pathway including the cytokines pro-interleukin (IL)-1β and pro-IL-18 are also transcriptionally upregulated. Noncanonical activation of procaspase-4 and -5 can also take place through their binding to lipopolysaccharide (LPS) in the cytoplasm. Once activated, inflammatory caspases cleave and activate GSDMD as noted above and process pro-IL-1β and pro-IL-18 to their mature forms, which escape through GSDMD-generated pores. Interaction of NLRP3 with mitochondrial antiviral signaling protein (MAVS) and cardiolipin facilitate inflammasome assembly and activation.
As noted, inflammatory caspases cleave GSDMD to GSDMD-NT (230, 439), although caspase-8 may also contribute in certain contexts (370). Activation of procaspase-1 occurs in multiprotein complexes known as inflammasomes. Canonical inflammasomes respond to diverse pathogen and host-derived danger signals, and typically consist of a pattern recognition receptor (PRR), which detects PAMPs and danger-associated molecular patterns (DAMPs), an adapter protein, and procaspase-1. Classic PRRs include NOD-like receptor (NLR) family members, the most well studied of which is NLR pyrin domain (PYD) containing receptor 3 (NLRP3). In the NLRP3 inflammasome, NLRP3 recruits the adapter protein apoptosis-associated specklike protein containing a CARD (ASC) through interactions involving the PYD in each protein, thereby providing a nucleation platform for ASC filament formation (296). Filamentous ASC “specks,” in turn, facilitate the recruitment and activation of caspase-1 through CARD-CARD interactions (256, 491). Noncanonical caspase activation may take place through the binding of gram-negative bacteria-derived lipopolysaccharide (LPS) to procaspases-4, -5, or -11 in the cytosol, which then triggers cleavage and activation (231, 440). Active caspase-1 processes pro-IL-1β and pro-IL-18 into mature IL-1β and IL-18 and promotes plasma membrane rupture via GSDMD culminating in lytic cell death and a pro-inflammatory milieu. Caspase-11 is sufficient to cleave GSDMD and induce cell death but does not directly process pro-IL-1β or pro-IL-18.
Inflammasome assembly depends on a two-step process that involves priming and activation. Priming occurs in response to receptor activation including engagement of TLR2, -3, or -4; TNFR1; or IL-1 receptor. Priming can be transcriptionally independent or dependent. Nontranscriptional, or early, priming occurs rapidly and transiently (10 min to 1 h after stimulation) as a result of posttranslational modifications of NLRP3. These include phosphorylation (451, 452, 457), ubiquitination (176, 385, 450, 536), and nitrosylation (188), events that are regulated proximally by the adapter proteins myeloid differentiation primary response 88 (MyD88), IL-1 receptor-associated kinase 1 (IRAK-1), and TRIF (126, 202, 217, 275, 393). Conversely, transcriptional priming takes longer (several hours) and is mediated by NF-κB, which elicits increased de novo expression of certain cytokines (pro-IL-1β, pro-IL-18) and inflammasome components (NLRP3, ASC, procaspase-1) that normally have low baseline expression (19, 170).
The intracellular location and precise mechanisms leading to inflammasome activation are still largely undefined. Studies in macrophages have demonstrated that mitochondria serve as nodes for canonical NLRP3 inflammasome assembly and signaling. In response to stimuli that trigger inflammasome activation, NLRP3 is recruited to mitochondria through an interaction with an OMM adapter referred to as mitochondrial anti-viral signaling protein (MAVS) (374, 458). The mitochondrial membrane component cardiolipin also binds to NLRP3 and may facilitate its subcellular translocation (208). Increases in mitochondrial-derived ROS and the oxidation of liberated mitochondrial DNA (mtDNA) from dysfunctional or damaged mitochondria drive assembly and activation of the NLRP3 inflammasome (343, 441, 565, 568). Oxidized mtDNA can also directly bind to NLRP3 to promote inflammasome function and subsequent caspase-1 activation (441). Interestingly, mitophagy is also stimulated during this process and regulates caspase-1 activity and inflammatory cytokine processing through the clearance of damaged mitochondria, thereby providing an important negative feedback mechanism (343, 566). Inflammasomes themselves can also be targeted for autophagic degradation, thereby promoting resolution of the inflammatory response (437). Perhaps, not surprisingly, inhibition of autophagy has been shown to promote inflammasome activation (343). While the role of these mitochondrial factors in modulating NLRP3 inflammasome activity has been demonstrated primarily in immune cells, their necessity in cardiac cell types, including cardiomyocytes, requires further investigation.
During I/R or non-reperfused MI, the expression of canonical inflammasome components (including ASC and NLRP3) is upregulated, as are the activation of caspase-1 and the secretion of pro-inflammatory cytokines IL-1β and IL-18 (229, 319, 417). However, the cell types in which inflammasome assembly and resulting cytokine processing take place remain unclear. In fact, it appears that inflammasome assembly is significantly more robust in non-cardiomyocytes (perhaps fibroblasts) and bone marrow-derived cells that are recruited into the heart during infarction than in cardiomyocytes themselves (229), although there is evidence that inflammasomes also assemble in cardiomyocytes to a more limited extent (319, 459, 529). Adoptive transfer experiments using ASC −/− mice as donors or recipients demonstrated that loss of inflammasome function in either the bone marrow compartment or in non-cardiomyocyte cardiac resident cells is sufficient to protect the heart from I/R. This suggests that inflammasome function is needed in both compartments for full infarction to take place (229) (TABLE 3). Cardiomyocyte-specific transgenic overexpression of procaspase-1 demonstrated that high levels of this inflammatory caspase are sufficient to activate caspase-3 (possibly an overexpression effect) and exacerbate apoptosis and infarct size in response to I/R (467), as well as under basal conditions, resulting in heart failure (317). Interestingly, however, this occurs in the absence of IL-1β and IL-18 maturation, despite the high levels of caspase-1 transgene expression in cardiomyocytes (317). This perhaps reflects sluggish inflammasome assembly in this cell type. Conversely, global deletion of procaspase-1 or ASC reduces infarct size following I/R (229), and global deletion of procaspase-1 ameliorates post-MI heart failure (317).
Since global knockout mice were used in all of these studies, however, it cannot be concluded that these effects are mediated by loss of caspase-1 in cardiomyocytes, especially in light of the results described above demonstrating that inflammasome assembly is more robust in non-cardiomyocytes (229). Targeting components of the inflammasome machinery using cell type-specific deletion approaches would help to clarify this issue. An important complementary approach would be to assess the effects of GSDMD ablation in various cardiac cell types.
4. Parthanatos
Parthanatos is initiated by, and dependent on, the hyperactivation of PARP-1, the subsequent accumulation of PAR polymers in the cytosol, and AIF-dependent DNA fragmentation (121). In addition to these biochemical signatures, parthanatos is distinguished from apoptosis by its lack of inter-nucleosomal DNA fragmentation and apoptotic bodies and from necrosis by the absence of cellular swelling (12, 552, 553). The prototypical stimulus for PARP-1 activation is DNA damage. However, oxidative stress, hypoxia, inflammatory stimuli, and excitotoxcity (in neurons) can also promote PARP-1 activation (70, 78, 121, 560). During parthanatos, elevated levels of PAR polymers precipitate the cleavage and release of AIF from mitochondria through mechanisms that remain incompletely understood but require calpains, BID, and BAX (42). The direct binding of AIF and PAR polymers may also facilitate this process (334, 513, 553). AIF, which lacks nuclease function, mediates large-scale DNA fragmentation and chromatin condensation independently of caspases or EndoG (307, 505, 513, 534). Instead, AIF binds and recruits MIF, a cytokine recently demonstrated to have nuclease activity, to the nucleus to execute DNA fragmentation (511). In addition, the excessive generation of PAR polymers by PARP-1 simultaneously depletes the cell of NAD+ and ATP, as discussed in section II, resulting in energetic collapse that is also thought to contribute to cytotoxicity and may provide an amplification loop for necrotic cell death (121).
Reperfusion of the infarcted myocardium generates a burst of reactive oxygen and nitrogen species that can promote the activity of PARP-1 (160, 297). Indeed, elevated PARP-1 expression and activation have been observed in rodent hearts after MI and failing human myocardium (269, 378, 379, 484). By utilizing global Parp-1 knockout mice, several independent groups have demonstrated that loss of PARP-1 confers cardioprotection against I/R injury. Isolated, perfused Parp-1 −/− hearts had more viable myocardium, improved contractility, and higher NAD+ content compared with wild-type counterparts following I/R (165, 378, 567) (TABLE 3). Consistent with these findings, Parp-1 −/− mice exhibited reduced infarct size and lower serum creatine kinase levels compared with wild-type mice following I/R in vivo (543, 573). With the use of an isolated, perfused rat heart model, treatment with a pharmacological inhibitor of PARP-1 afforded improved functional recovery of the reperfused myocardium (98, 567). Administration of a PARP-1 inhibitor in vivo also conferred a cardioprotective benefit in response to I/R in the rat. Inhibitor treatment reduced infarct size and attenuated systolic dysfunction, largely mirroring results obtained using Parp-1 −/− mice (269, 517, 572). Parp-1 −/− mice were also protected from pressure overload-induced cardiac dysfunction and pathological remodeling, although the underlying mechanism was not defined (379). Importantly, myocardial apoptosis (as determined by TUNEL and DNA laddering) was reduced in Parp-1 −/− mice following I/R (573). However, established features of parthanatos were not rigorously examined in these studies, as most of this work preceded the definition of parthanatos as a regulated cell death program. Therefore, whether parthanatos per se occurs in the injured and/or failing heart and whether specific inhibition of this cell death program provides benefit remain unknown. To this point, two studies using Mif −/− mice demonstrated that loss of endogenous MIF paradoxically exacerbated infarct size and further suppressed functional recovery following I/R (324, 387). Moreover, whether parthanatos occurs within, and kills, cardiomyocytes has not been investigated, although forced expression of PARP-1 in neonatal rat cardiomyocytes was shown to reduce cell viability (the type of death and underlying mechanism were not explored) (379). Addressing these points remains critical to our basic understanding of parthanatos in heart disease. Nevertheless, based on these promising preclinical results, a phase I clinical trial (safety and dosing) of the PARP-1 inhibitor INO-1001 was performed in 40 patients with STEMI undergoing percutaneous coronary intervention (PCI) (330). INO-1001 reduced serum PARP-1 activity and inflammatory markers, although also resulted in elevation of some liver transaminases. For unclear reasons, clinical testing never progressed to phase 2.
5. Immunogenic cell death
Immunogenic cell death (ICD) is a regulated process in which dying cells are able to initiate an immune response that kills other cells expressing the same antigens (140, 142, 143). This process is initiated when the dying cells expose or release DAMPs. DAMPs include endogenous cellular proteins, nucleic acids, and small molecules such as the ER protein calreticulin, the chromatin binding protein high-mobility group box 1 (HMGB1), type I IFNs, annexin A1, nucleic acids, and ATP. DAMPs bind to PRRs on or within antigen presenting cells (APCs), such as dendritic cells. The APCs subsequently internalize the dead cell corpses providing antigens that processed and displayed on the surface of APCs as peptides bound to major histocompatibility complex (MHC) molecules. Binding of this complex to an appropriate T cell receptor, along with a costimulus provided by an interaction of B7-1 (CD80) or B7-2 (CD86) on the APC to CD28 on the T cell, stimulates T cell proliferation and differentiation. This process is referred to as T cell activation (253). Activated T cells are then recruited and kill target cells (e.g., cancer cells) expressing the same antigens as those expressed by the dying cells that initiated this process. This specificity is provided by an interaction between peptide MHC on the target cells with the T cell receptor.
Conversely, T cell activation is opposed by a variety of mechanisms (20). These include inhibition mediated by cytotoxic T lymphocyte-associated protein 4 (CTLA-4; CD152) and programmed cell death protein 1 (PD-1; CD279), both proteins resident on the surface of T cells (400). [It should be noted that PD-1, despite its name, is not primarily involved in cell death (4).] While some functional overlap exists, CTLA-4 restrains T cells primarily in secondary lymphoid organs, and PD-1 exerts its effects mainly in peripheral tissues (522). Stimuli that activate T cells induce CTLA-4 trafficking to the cell surface allowing this protein to steal B7-1 and B7-2 away from binding to CD28, thereby depriving T cells of this activating costimulus (280, 281, 491). T cell-activating stimuli also induce the expression of PD-1. Ligands for PD-1 include the cell surface proteins programmed cell death ligand 1 (PD-L1; CD274), which is expressed on APCs and cells of multiple tissues, and PD-L2 (CD273), whose expression is more limited (103, 116, 133, 254, 255). PD-L1 or PD-L2 binding to PD-1 activates Src homology 2-containing phosphotyrosine phosphatase (SHP2), which inhibits signaling from the T cell receptor and CD28 (201, 550). In addition, the expression of PD-L1 and PD-L2 are induced by IFN-γ secreted by activated T cells, which provides a negative feedback loop that restrains T cell activation to protect peripheral tissues from damage (133, 145, 254). In addition to these canonical mechanisms, further complexity continues to emerge in this intensively studied field (400, 522).
Elucidation of the CTLA-4 and PD-1 pathways provided the cornerstone for the development of immune checkpoint inhibitor (ICI) therapies for cancer, arguably the most dramatic advance in cancer treatment in the past decade (400). Although cancer cells use the mechanisms described above to activate immunogenic cell death, these cells often evade immune surveillance through a number of mechanisms (20, 141, 372, 399). For example, they can overexpress PD-L1 to prevent T cell activation. ICIs are monoclonal antibodies that bind CTLA-4 (ipilimumab), PD-1 (nivolumab, pembrolizumab, cemiplimab), or PD-L1 (atezolizumab, avelumab, durvalumab) and, in so doing, block these proteins from engaging in the interactions described above (400). The result is an unleashing of T cell activation that promotes cancer regression leading to an unprecedented extension of life in some of the most refractory and lethal malignancies.
As might be anticipated, ICIs can also induce immune-related damage to multiple tissues (215, 358, 518). In particular, myocarditis has been reported (197, 214, 301, 333, 414). The median time of onset following initiation of therapy has been reported to be 27–30 days, but with variability (333, 414). Clinical manifestations have included heart block, refractory ventricular tachycardia, and heart failure. A retrospective analysis of 101 cases of severe ICI-associated myocarditis indicated an overall mortality of 46%, rising to 67% when two ICIs were employed (333). ICI-associated myocarditis is characterized by lymphocytic and macrophage infiltration involving both the conduction system and cardiac muscle (214). Elevations in serum troponin levels have been noted consistent with cardiomyocyte necrosis (214, 301). Deep sequencing of the T cell receptor of two patients suggested that infiltrates in heart, skeletal muscle, and the cancer showed clonal expansion of similar T cell populations (214). More recently pericarditis and vasculitis have also been associated with ICIs (414).
The occurrence of ICI-associated myocarditis is not unexpected based on studies in mice. For example, germline deletion of Pdcd1, encoding PD-1, in BALB/c mice caused myocarditis and dilated cardiomyopathy (357). Pathogenesis in this case appears to have been driven by an autoantibody against cardiac troponin I (365). Pdcd1 knockout has also been reported to exacerbate myosin peptide-induced myocarditis, and this was mediated by CD4+ T cells (476). In addition, adoptive transfer experiments showed that deletion of Pdcd1 in CD8+ T cells exacerbated myocarditis induced by ovalbumin (476). Second, germline combined knockout of CD274 (encoding PD-L1) and CD273 (encoding PD-L2) also exacerbated the severity of ovalbumin-induced myocarditis (157). This outcome was phenocopied by administration of a blocking antibody against mouse PD-L1 (157). Finally, germline knockout of Ctla4 caused a lethal lymphoproliferative syndrome, which included myocarditis (48, 237, 481, 516).
The frequency of ICI-associated myocarditis is quite low, estimated to be only ~1% (301). While one explanation for this low occurrence rate is likely under-reporting of this new syndrome, another is the basal inhibition of immune mechanisms in the myocardium (158). One inhibitory mechanism appears to involve induction of PD-L1 expression in cardiomyocytes and microvascular endothelial cells in the heart in response to several stress stimuli. Despite the currently reported low frequency of ICI-associated myocarditis, we suspect that this syndrome will become more common as ICIs become even more widely employed and used in combination with other cancer therapies that promote DAMP release from both cancer cells as intended and from cardiac cells as an untoward effect (e.g., doxorubicin and radiation; Ref. 107).
The discussion thus far has focused on how ICIs unleash T cell activation, which can promote cancer regression and induce autoimmune tissue damage. However, we have not yet considered the mechanisms by which cytotoxic lymphocytes kill cells. In cancer contexts, two mechanisms have been demonstrated (304). In the first, the binding of death ligands on the plasma membrane of (FasL) or secreted from (TRAIL, TNF-α) cytotoxic lymphocytes can induce cancer cell death through binding to their cognate death receptors on the cancer cell, as we have previously discussed. The second killing mechanism involves perforin-1 and granzyme B. Cytoplasmic granules containing perforin-1 and granzyme B are released from cytotoxic lymphocytes in a directional manner such that they enter the cleft between the T cell and the cancer cell (immunological synapse) (501). Perforin-1 creates pores in the cancer cell plasma membrane allowing entry of granzyme B. Granzyme B is a serine protease that hydrolyzes peptide linkages following aspartic acid residues in the preferred motif IEXD↓XG. This bears some similarity to preferred motif of caspase-8 [(I/L/V)EXD↓] (177, 480). Consistent with this, granzyme B is able to cleave and activate caspase-8 substrates BID (18, 515) and procaspase-3 (75, 155, 222, 318). While these events are suggestive, the mechanisms by which granzyme B actually kills cells is more complex and differs between humans and rodents (222). Human granzyme B can induce cell death by cleaving BID leading to BAX/BAK-dependent MOMP, cytochrome c release, and procaspase-3 activation (186, 380). Mouse granzyme B, on the other hand, can directly cleave and activate procaspase-3 (222). In addition, granzyme B can also kill cells lacking BID or BAX/BAK, suggesting the existence of other cell death pathways (479). One involves granzyme B-mediated cleavage of mitochondrial complex I components NDUFS1, NDUFS2, and NDUFV1 resulting in a ROS-dependent form of cell death, which could be necrotic as well as apoptotic (59, 210). It is possible that different mechanisms may be activated by granzyme B depending on which death options remain available in the context of the genetic mutations in a given cancer cell.
In contrast to cancer cells, the extent to which ICI-activated T cells kill cardiomyocytes and other heart cells is unclear. While ICIs can result in inflammatory infiltrates involving both cardiomyocytes and the conduction system and while there is evidence of cardiomyocyte necrosis (troponin leak), several key issues remain unclear: 1) To what extent does cardiac cell death contribute to the pathogenesis of ICI-associated myocarditis? 2) Which cell death programs are involved? 3) Are the mechanisms by which T cells kill cardiac cells the same or different from those utilized for cancer cells? As ICIs become even more widely deployed, answers to these questions will be critical.
Finally, it is worth considering whether immunogenic cell death, in the absence of ICIs, may contribute to cardiomyocyte loss during MI or heart failure. This may seem unlikely given the baseline inhibition of immune mechanisms in the myocardium and the fact that this inhibition appears to intensify under stress conditions (158). In addition, involvement of immunogenic cell death may appear “unnecessary” given the plethora of cardiomyocyte-intrinsic death programs described in this review. Nevertheless, further investigation will be needed to test this possibility.
IV. CONCLUSIONS AND THERAPEUTIC CONSIDERATIONS
A. Summary of the Roles of Regulated Cell Death in MI and Heart Failure
The most important conclusion from the above studies is that regulated forms of cell death contribute significantly to the pathogenesis of both MI with reperfusion (I/R) and heart failure (specifically HFrEF). Some caveats need to be emphasized, however. First, while heart samples from patients with these syndromes show various markers confirming the occurrence of regulated cell death programs, the genetic and pharmacological studies establishing cause and effect were performed in animal models. Second, the primacy of cell death in pathogenesis differs greatly between MI and heart failure. During acute MI, cardiomyocyte death is the central event in pathogenesis. Acute loss of these cells in the infarct zone drives subsequent adverse remodeling in the remaining myocardium leading to heart failure. Importantly, the studies presented above show that much of this acute cell death occurs through regulated death programs and, therefore, may be amenable to therapeutic manipulation. In contrast, while levels of cardiomyocyte death are substantially elevated in heart failure, their absolute magnitude is quite low compared with acute MI. Genetic studies in mice, however, again show that this cell death occurs through regulated programs and, in aggregate, contributes to heart failure progression. Nevertheless, cell death is only one component in the pathogenesis of heart failure as dysfunction of viable cardiomyocytes is also important.
What cell death programs are most important in I/R and heart failure? Recent data prompt reconsideration of the relative contributions of apoptosis and necrosis in I/R. Early studies suggested that apoptosis was the primary form of cell death in this syndrome. In some cases, this conclusion was based on genetic manipulations that were thought at the time to impact only apoptotic cell death but were later recognized to also affect necrosis. Raising further questions concerning a role for apoptosis in I/R are data suggesting that caspase-dependent cell death is less robust in adult as compared with fetal/neonatal cardiomyocytes (415, 416, 421). What do we know now? While current data clearly establish a role for non-apoptotic cell death programs in I/R, we remain uncertain whether cardiomyocyte apoptosis plays a role. Given the temporal and spatial complexity of cell death during I/R, we feel that further studies to examine this issue are needed. In the interim, however, we suggest that authors employ the term cardiomyocyte death in the context of I/R rather than the term cardiomyocyte apoptosis.
What non-apoptotic cell death programs mediate cardiomyocyte loss during I/R? The studies presented above demonstrate a role for both mitochondria-mediated necrosis and necroptosis. In addition, emerging data implicate ferroptosis, although it remains unclear whether this program is functioning as an independent cell death modality or an amplification loop for the necrotic cell death. Regardless, the molecules that mediate ferroptosis may provide new therapeutic targets. PARP-1-dependent processes also appear important in I/R. Although we discussed these in the context of the cell death program parthanatos, the significance of PARP-1 activation may be more related to its depletion of NAD+ and disruption of cellular energetics. The data regarding autosis in ischemic neuronal injury are intriguing, and encourage further study of this form of autophagy-dependent cell death in myocardial I/R. Pyroptosis may also contribute to I/R, but the major challenge is in understanding the cell types in which it is taking place. Current data suggest that pyroptosis operates primarily in non-cardiomyocytes, bone marrow-derived cells recruited to the heart during MI and perhaps cardiac fibroblasts, but further study is needed.
What cell death programs contribute to the pathogenesis of heart failure? Both apoptotic and necrotic cardiomyocyte death have been implicated, but their relative contributions remain unclear. Very limited information is available regarding the roles of other cell death programs in heart failure.
B. Therapeutic Considerations
The first issue is which cardiac syndromes provide the optimal targets for anti-cell death therapies. We would argue that those requiring short-term inhibition of cell death may be the most logical starting points because they limit the possibility of undesirable outcomes including the initiation/progression of cancer and interference with antiviral defenses. The most common and important acute indication is I/R in the setting of MI or possibly cardiac surgery including transplantation. A second important short-term indication may be in the prevention of cardiotoxicities related to cancer treatments. These often involve a cell death component and can result from conventional chemotherapy, targeted therapies, and radiation (331, 332). An additional advantage of targeting cell death in the contexts of cardiac surgery and cancer treatments is that, unlike MI, these are scheduled events allowing a therapy to be initiated at or before death stimuli. What about targeting cell death in nonacute conditions such as heart failure? Given the importance of this syndrome and its residual mortality despite current treatments, heart failure may well be a future candidate for therapies that inhibit cardiomyocyte death. While the necessity for ongoing cell death inhibition raises the concerns noted above, the lethality of heart failure may justify such an approach. Risk-benefit analyses are an important consideration for all medical treatments.
Although an effective therapy for I/R would be impactful, this has traditionally been a very difficult problem. How can cell death be inhibited in I/R? Based on the data presented, targeting necrosis in the death receptor and mitochondrial pathways would likely be the highest priorities, although inhibition of ferroptosis, PARP-1 activation, and autosis might also be beneficial. The most promising approaches in the death receptor pathway involve inhibition of RIPK1 and RIPK3, and the latter may have the advantage of possessing a wider spectrum of relevant downstream targets (351, 563). Several possible approaches exist to inhibiting necrosis in the mitochondrial pathway. The most obvious is to inhibit mPTP opening. As discussed, CsA showed initial promise in a small clinical trial, but later failed in further testing (69, 381). While there may be multiple explanations for this result (278), one possibility is that CsA is not the optimal drug to antagonize cyclophilin D and mPTP opening. Another possibility is that additional positive regulators of mPTP may also require inactivation. An understanding of the proteins that constitute mPTP could provide insights into this question as well as reveal novel drug targets. Another potential target in the mitochondrial pathway is BAX. In fact, highly selective small molecule inhibitors of BAX have recently been reported (146), which have the theoretical advantage of inhibiting both necrotic and apoptotic cell death.
GRANTS
This work was supported by National Institutes of Health Grants R01HL127339 and R15HL135726 (to D. P. Del Re), R01HL116507 (to Q. Liu), as well as R01HL128071, R01 HL130861, and R01HL138475 (to R. N. Kitsis); Department of Defense Grant PR151134P1 (to R. N. Kitsis); American Heart Association Grants 15PRE25080032 (to D. Amgalan) as well as 15CSA26240000 and 18SRG34280018 (to R. N. Kitsis); Fondation Leducq (to R. N. Kitsis); Deutsche Forschungsgemeinschaft Grant EXC 306 (to A. Linkermann); and research grants from the Else Kröner-Fresenius Stiftung (to A. Linkermann).
DISCLOSURES
A. Linkermann has received research grants from Pfizer, Novartis, Fresenius Medical Care, and the Else Kröner-Fresenius Stiftung; has material transfer agreements with Genentech, Glaxo Smith Kline, and Apogenix; and has received honoraria and/or travel grants from Astellas, Otsuka, Genentech, Alexion, and Tekmira. R.N. Kitsis is co-founder of Aspida Therapeutics Inc. and a consultant for Amaron Bio. No conflicts of interest, financial or otherwise, are declared by the other authors.
ACKNOWLEDGMENTS
D. P. Del Re and D. Amgalan contributed equally to this work.
All images were created with BioRender.com.
Address for reprint requests and other correspondence: R. N. Kitsis, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY 10461 (e-mail: richard.kitsis@einstein.yu.edu).
REFERENCES
- 1.Abelson S, Collord G, Ng SWK, Weissbrod O, Mendelson Cohen N, Niemeyer E, Barda N, Zuzarte PC, Heisler L, Sundaravadanam Y, Luben R, Hayat S, Wang TT, Zhao Z, Cirlan I, Pugh TJ, Soave D, Ng K, Latimer C, Hardy C, Raine K, Jones D, Hoult D, Britten A, McPherson JD, Johansson M, Mbabaali F, Eagles J, Miller JK, Pasternack D, Timms L, Krzyzanowski P, Awadalla P, Costa R, Segal E, Bratman SV, Beer P, Behjati S, Martincorena I, Wang JCY, Bowles KM, Quirós JR, Karakatsani A, La Vecchia C, Trichopoulou A, Salamanca-Fernández E, Huerta JM, Barricarte A, Travis RC, Tumino R, Masala G, Boeing H, Panico S, Kaaks R, Krämer A, Sieri S, Riboli E, Vineis P, Foll M, McKay J, Polidoro S, Sala N, Khaw K-T, Vermeulen R, Campbell PJ, Papaemmanuil E, Minden MD, Tanay A, Balicer RD, Wareham NJ, Gerstung M, Dick JE, Brennan P, Vassiliou GS, Shlush LI. Prediction of acute myeloid leukaemia risk in healthy individuals. Nature 559: 400–404, 2018. doi: 10.1038/s41586-018-0317-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Acehan D, Jiang X, Morgan DG, Heuser JE, Wang X, Akey CW. Three-dimensional structure of the apoptosome: implications for assembly, procaspase-9 binding, and activation. Mol Cell 9: 423–432, 2002. doi: 10.1016/S1097-2765(02)00442-2. [DOI] [PubMed] [Google Scholar]
- 3.Adams JW, Sakata Y, Davis MG, Sah VP, Wang Y, Liggett SB, Chien KR, Brown JH, Dorn GW II. Enhanced Galphaq signaling: a common pathway mediates cardiac hypertrophy and apoptotic heart failure. Proc Natl Acad Sci USA 95: 10140–10145, 1998. doi: 10.1073/pnas.95.17.10140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Agata Y, Kawasaki A, Nishimura H, Ishida Y, Tsubat T, Yagita H, Honjo T. Expression of the PD-1 antigen on the surface of stimulated mouse T and B lymphocytes. Int Immunol 8: 765–772, 1996. doi: 10.1093/intimm/8.5.765. [DOI] [PubMed] [Google Scholar]
- 5.Aggarwal BB. Signalling pathways of the TNF superfamily: a double-edged sword. Nat Rev Immunol 3: 745–756, 2003. doi: 10.1038/nri1184. [DOI] [PubMed] [Google Scholar]
- 6.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Alvarez SE, Harikumar KB, Hait NC, Allegood J, Strub GM, Kim EY, Maceyka M, Jiang H, Luo C, Kordula T, Milstien S, Spiegel S. Sphingosine-1-phosphate is a missing cofactor for the E3 ubiquitin ligase TRAF2. Nature 465: 1084–1088, 2010. doi: 10.1038/nature09128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Alvarez-Diaz S, Dillon CP, Lalaoui N, Tanzer MC, Rodriguez DA, Lin A, Lebois M, Hakem R, Josefsson EC, O’Reilly LA, Silke J, Alexander WS, Green DR, Strasser A. The Pseudokinase MLKL and the Kinase RIPK3 Have Distinct Roles in Autoimmune Disease Caused by Loss of Death-Receptor-Induced Apoptosis. Immunity 45: 513–526, 2016. doi: 10.1016/j.immuni.2016.07.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Amgalan D, Chen Y, Kitsis RN. Death Receptor Signaling in the Heart: Cell Survival, Apoptosis, and Necroptosis. Circulation 136: 743–746, 2017. doi: 10.1161/CIRCULATIONAHA.117.029566. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Amgalan D, Pekson R, Kitsis RN. Troponin release following brief myocardial ischemia: apoptosis versus necrosis. JACC Basic Transl Sci 2: 118–121, 2017. doi: 10.1016/j.jacbts.2017.03.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Anding AL, Baehrecke EH. Autophagy in Cell Life and Cell Death. Curr Top Dev Biol 114: 67–91, 2015. doi: 10.1016/bs.ctdb.2015.07.012. [DOI] [PubMed] [Google Scholar]
- 12.Andrabi SA, Kim NS, Yu SW, Wang H, Koh DW, Sasaki M, Klaus JA, Otsuka T, Zhang Z, Koehler RC, Hurn PD, Poirier GG, Dawson VL, Dawson TM. Poly(ADP-ribose) (PAR) polymer is a death signal. Proc Natl Acad Sci USA 103: 18308–18313, 2006. doi: 10.1073/pnas.0606526103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Aoyagi T, Kusakari Y, Xiao CY, Inouye BT, Takahashi M, Scherrer-Crosbie M, Rosenzweig A, Hara K, Matsui T. Cardiac mTOR protects the heart against ischemia-reperfusion injury. Am J Physiol Heart Circ Physiol 303: H75–H85, 2012. doi: 10.1152/ajpheart.00241.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Ashkenazi A, Dixit VM. Death receptors: signaling and modulation. Science 281: 1305–1308, 1998. doi: 10.1126/science.281.5381.1305. [DOI] [PubMed] [Google Scholar]
- 15.Baba Y, Higa JK, Shimada BK, Horiuchi KM, Suhara T, Kobayashi M, Woo JD, Aoyagi H, Marh KS, Kitaoka H, Matsui T. Protective effects of the mechanistic target of rapamycin against excess iron and ferroptosis in cardiomyocytes. Am J Physiol Heart Circ Physiol 314: H659–H668, 2018. doi: 10.1152/ajpheart.00452.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Baines CP, Gutiérrez-Aguilar M. The still uncertain identity of the channel-forming unit(s) of the mitochondrial permeability transition pore. Cell Calcium 73: 121–130, 2018. doi: 10.1016/j.ceca.2018.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Baines CP, Kaiser RA, Purcell NH, Blair NS, Osinska H, Hambleton MA, Brunskill EW, Sayen MR, Gottlieb RA, Dorn GW, Robbins J, Molkentin JD. Loss of cyclophilin D reveals a critical role for mitochondrial permeability transition in cell death. Nature 434: 658–662, 2005. doi: 10.1038/nature03434. [DOI] [PubMed] [Google Scholar]
- 18.Barry M, Heibein JA, Pinkoski MJ, Lee SF, Moyer RW, Green DR, Bleackley RC. Granzyme B short-circuits the need for caspase 8 activity during granule-mediated cytotoxic T-lymphocyte killing by directly cleaving Bid. Mol Cell Biol 20: 3781–3794, 2000. doi: 10.1128/MCB.20.11.3781-3794.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Bauernfeind FG, Horvath G, Stutz A, Alnemri ES, MacDonald K, Speert D, Fernandes-Alnemri T, Wu J, Monks BG, Fitzgerald KA, Hornung V, Latz E. Cutting edge: NF-kappaB activating pattern recognition and cytokine receptors license NLRP3 inflammasome activation by regulating NLRP3 expression. J Immunol 183: 787–791, 2009. doi: 10.4049/jimmunol.0901363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Baumeister SH, Freeman GJ, Dranoff G, Sharpe AH. Coinhibitory Pathways in Immunotherapy for Cancer. Annu Rev Immunol 34: 539–573, 2016. doi: 10.1146/annurev-immunol-032414-112049. [DOI] [PubMed] [Google Scholar]
- 21.Bernardi P. Why F-ATP Synthase Remains a Strong Candidate as the Mitochondrial Permeability Transition Pore. Front Physiol 9: 1543, 2018. doi: 10.3389/fphys.2018.01543. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Bertrand MJ, Milutinovic S, Dickson KM, Ho WC, Boudreault A, Durkin J, Gillard JW, Jaquith JB, Morris SJ, Barker PA. cIAP1 and cIAP2 facilitate cancer cell survival by functioning as E3 ligases that promote RIP1 ubiquitination. Mol Cell 30: 689–700, 2008. doi: 10.1016/j.molcel.2008.05.014. [DOI] [PubMed] [Google Scholar]
- 24.Bhuiyan MS, Fukunaga K. Inhibition of HtrA2/Omi ameliorates heart dysfunction following ischemia/reperfusion injury in rat heart in vivo. Eur J Pharmacol 557: 168–177, 2007. doi: 10.1016/j.ejphar.2006.10.067. [DOI] [PubMed] [Google Scholar]
- 25.Birge RB, Ucker DS. Innate apoptotic immunity: the calming touch of death. Cell Death Differ 15: 1096–1102, 2008. doi: 10.1038/cdd.2008.58. [DOI] [PubMed] [Google Scholar]
- 26.Biton S, Ashkenazi A. NEMO and RIP1 control cell fate in response to extensive DNA damage via TNF-α feedforward signaling. Cell 145: 92–103, 2011. doi: 10.1016/j.cell.2011.02.023. [DOI] [PubMed] [Google Scholar]
- 27.Blazek AD, Paleo BJ, Weisleder N. Plasma Membrane Repair: A Central Process for Maintaining Cellular Homeostasis. Physiology (Bethesda) 30: 438–448, 2015. doi: 10.1152/physiol.00019.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Boatright KM, Renatus M, Scott FL, Sperandio S, Shin H, Pedersen IM, Ricci JE, Edris WA, Sutherlin DP, Green DR, Salvesen GS. A unified model for apical caspase activation. Mol Cell 11: 529–541, 2003. doi: 10.1016/S1097-2765(03)00051-0. [DOI] [PubMed] [Google Scholar]
- 29.Bogan KL, Brenner C. Nicotinic acid, nicotinamide, and nicotinamide riboside: a molecular evaluation of NAD+ precursor vitamins in human nutrition. Annu Rev Nutr 28: 115–130, 2008. doi: 10.1146/annurev.nutr.28.061807.155443. [DOI] [PubMed] [Google Scholar]
- 30.Boldin MP, Goncharov TM, Goltseve YV, Wallach D. Involvement of MACH, a novel MORT1/FADD-interacting protease, in Fas/APO-1- and TNF receptor-induced cell death. Cell 85: 803–815, 1996. doi: 10.1016/S0092-8674(00)81265-9. [DOI] [PubMed] [Google Scholar]
- 31.Bolli R, Becker L, Gross G, Mentzer R Jr, Balshaw D, Lathrop DA; NHLBI Working Group on the Translation of Therapies for Protecting the Heart from Ischemia . Myocardial protection at a crossroads: the need for translation into clinical therapy. Circ Res 95: 125–134, 2004. doi: 10.1161/01.RES.0000137171.97172.d7. [DOI] [PubMed] [Google Scholar]
- 32.Bonaventura A, Montecucco F, Dallegri F, Carbone F, Lüscher TF, Camici GG, Liberale L. Novel findings in neutrophil biology and their impact on cardiovascular disease. Cardiovasc Res 115: 1266–1285, 2019. doi: 10.1093/cvr/cvz084. [DOI] [PubMed] [Google Scholar]
- 33.Bonora M, Bononi A, De Marchi E, Giorgi C, Lebiedzinska M, Marchi S, Patergnani S, Rimessi A, Suski JM, Wojtala A, Wieckowski MR, Kroemer G, Galluzzi L, Pinton P. Role of the c subunit of the FO ATP synthase in mitochondrial permeability transition. Cell Cycle 12: 674–683, 2013. doi: 10.4161/cc.23599. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Borlaug BA, Redfield MM. Diastolic and systolic heart failure are distinct phenotypes within the heart failure spectrum. Circulation 123: 2006–2013, 2011. doi: 10.1161/CIRCULATIONAHA.110.954388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Bouchier-Hayes L, Green DR. Caspase-2: the orphan caspase. Cell Death Differ 19: 51–57, 2012. doi: 10.1038/cdd.2011.157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Boyd JM. Adenovirus E1B 19 kDa and Bcl-2 proteins interact with a common set of cellular proteins. Cell 79: 341–351, 1994. doi: 10.1016/0092-8674(94)90202-X. [DOI] [PubMed] [Google Scholar]
- 37.Brenner D, Blaser H, Mak TW. Regulation of tumour necrosis factor signalling: live or let die. Nat Rev Immunol 15: 362–374, 2015. doi: 10.1038/nri3834. [DOI] [PubMed] [Google Scholar]
- 38.Brocheriou V, Hagège AA, Oubenaïssa A, Lambert M, Mallet VO, Duriez M, Wassef M, Kahn A, Menasché P, Gilgenkrantz H. Cardiac functional improvement by a human Bcl-2 transgene in a mouse model of ischemia/reperfusion injury. J Gene Med 2: 326–333, 2000. doi:. [DOI] [PubMed] [Google Scholar]
- 39.Budd RC, Yeh WC, Tschopp J. cFLIP regulation of lymphocyte activation and development. Nat Rev Immunol 6: 196–204, 2006. doi: 10.1038/nri1787. [DOI] [PubMed] [Google Scholar]
- 40.Bulluck H, Rosmini S, Abdel-Gadir A, White SK, Bhuva AN, Treibel TA, Fontana M, Ramlall M, Hamarneh A, Sirker A, Herrey AS, Manisty C, Yellon DM, Kellman P, Moon JC, Hausenloy DJ. Residual Myocardial Iron Following Intramyocardial Hemorrhage During the Convalescent Phase of Reperfused ST-Segment-Elevation Myocardial Infarction and Adverse Left Ventricular Remodeling. Circ Cardiovasc Imaging 9: e004940, 2016. doi: 10.1161/CIRCIMAGING.116.004940. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Burchfield JS, Dong JW, Sakata Y, Gao F, Tzeng HP, Topkara VK, Entman ML, Sivasubramanian N, Mann DL. The cytoprotective effects of tumor necrosis factor are conveyed through tumor necrosis factor receptor-associated factor 2 in the heart. Circ Heart Fail 3: 157–164, 2010. doi: 10.1161/CIRCHEARTFAILURE.109.899732. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Cabon L, Galán-Malo P, Bouharrour A, Delavallée L, Brunelle-Navas MN, Lorenzo HK, Gross A, Susin SA. BID regulates AIF-mediated caspase-independent necroptosis by promoting BAX activation. Cell Death Differ 19: 245–256, 2012. doi: 10.1038/cdd.2011.91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Cai Z, Jitkaew S, Zhao J, Chiang HC, Choksi S, Liu J, Ward Y, Wu LG, Liu ZG. Plasma membrane translocation of trimerized MLKL protein is required for TNF-induced necroptosis. [Erratum in Nat Cell Biol 16: 200, 2014.] Nat Cell Biol 16: 55–65, 2014. doi: 10.1038/ncb2883. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Caporali A, Sala-Newby GB, Meloni M, Graiani G, Pani E, Cristofaro B, Newby AC, Madeddu P, Emanueli C. Identification of the prosurvival activity of nerve growth factor on cardiac myocytes. Cell Death Differ 15: 299–311, 2008. doi: 10.1038/sj.cdd.4402263. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Cassidy-Stone A, Chipuk JE, Ingerman E, Song C, Yoo C, Kuwana T, Kurth MJ, Shaw JT, Hinshaw JE, Green DR, Nunnari J. Chemical inhibition of the mitochondrial division dynamin reveals its role in Bax/Bak-dependent mitochondrial outer membrane permeabilization. Dev Cell 14: 193–204, 2008. doi: 10.1016/j.devcel.2007.11.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Catz SD, Johnson JL. Transcriptional regulation of bcl-2 by nuclear factor kappa B and its significance in prostate cancer. Oncogene 20: 7342–7351, 2001. doi: 10.1038/sj.onc.1204926. [DOI] [PubMed] [Google Scholar]
- 47.Chai J, Shiozaki E, Srinivasula SM, Wu Q, Dataa P, Alnemri ES, Shi Y. Structural basis of caspase-7 inhibition by XIAP [Correction in Cell 107: 409, 2001.]. Cell 104: 769–780, 2001. doi: 10.1016/S0092-8674(01)00272-0. [DOI] [PubMed] [Google Scholar]
- 48.Chambers CA, Cado D, Truong T, Allison JP. Thymocyte development is normal in CTLA-4-deficient mice. Proc Natl Acad Sci USA 94: 9296–9301, 1997. doi: 10.1073/pnas.94.17.9296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Chen G, Ray R, Dubik D, Shi L, Cizeau J, Bleackley RC, Saxena S, Gietz RD, Greenberg AH. The E1B 19K/Bcl-2-binding protein Nip3 is a dimeric mitochondrial protein that activates apoptosis. J Exp Med 186: 1975–1983, 1997. doi: 10.1084/jem.186.12.1975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Chen GY, Nuñez G. Sterile inflammation: sensing and reacting to damage. Nat Rev Immunol 10: 826–837, 2010. doi: 10.1038/nri2873. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Chen X, Li W, Ren J, Huang D, He WT, Song Y, Yang C, Li W, Zheng X, Chen P, Han J. Translocation of mixed lineage kinase domain-like protein to plasma membrane leads to necrotic cell death. Cell Res 24: 105–121, 2014. doi: 10.1038/cr.2013.171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Chen Y, Dorn GW II. PINK1-phosphorylated mitofusin 2 is a Parkin receptor for culling damaged mitochondria. Science 340: 471–475, 2013. doi: 10.1126/science.1231031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Chen Y, Lewis W, Diwan A, Cheng EH, Matkovich SJ, Dorn GW II. Dual autonomous mitochondrial cell death pathways are activated by Nix/BNip3L and induce cardiomyopathy. Proc Natl Acad Sci USA 107: 9035–9042, 2010. doi: 10.1073/pnas.0914013107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Chen Z, Chua CC, Ho YS, Hamdy RC, Chua BH. Overexpression of Bcl-2 attenuates apoptosis and protects against myocardial I/R injury in transgenic mice. Am J Physiol Heart Circ Physiol 280: H2313–H2320, 2001. doi: 10.1152/ajpheart.2001.280.5.H2313. [DOI] [PubMed] [Google Scholar]
- 55.Chinnaiyan AM, O’Rourke K, Tewari M, Dixit VM. FADD, a novel death domain-containing protein, interacts with the death domain of Fas and initiates apoptosis. Cell 81: 505–512, 1995. doi: 10.1016/0092-8674(95)90071-3. [DOI] [PubMed] [Google Scholar]
- 56.Chipuk JE, McStay GP, Bharti A, Kuwana T, Clarke CJ, Siskind LJ, Obeid LM, Green DR. Sphingolipid metabolism cooperates with BAK and BAX to promote the mitochondrial pathway of apoptosis. Cell 148: 988–1000, 2012. doi: 10.1016/j.cell.2012.01.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Chipuk JE, Moldoveanu T, Llambi F, Parsons MJ, Green DR. The BCL-2 family reunion. Mol Cell 37: 299–310, 2010. doi: 10.1016/j.molcel.2010.01.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Chiusolo V, Jacquemin G, Yonca Bassoy E, Vinet L, Liguori L, Walch M, Kozjak-Pavlovic V, Martinvalet D. Granzyme B enters the mitochondria in a Sam50-, Tim22- and mtHsp70-dependent manner to induce apoptosis. Cell Death Differ 24: 747–758, 2017. doi: 10.1038/cdd.2017.3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Cho Y, McQuade T, Zhang H, Zhang J, Chan FK. RIP1-dependent and independent effects of necrostatin-1 in necrosis and T cell activation. PLoS One 6: e23209, 2011. doi: 10.1371/journal.pone.0023209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Cho YS, Challa S, Moquin D, Genga R, Ray TD, Guildford M, Chan FK. Phosphorylation-driven assembly of the RIP1-RIP3 complex regulates programmed necrosis and virus-induced inflammation. Cell 137: 1112–1123, 2009. doi: 10.1016/j.cell.2009.05.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Chua CC, Gao J, Ho YS, Xiong Y, Xu X, Chen Z, Hamdy RC, Chua BH. Overexpression of IAP-2 attenuates apoptosis and protects against myocardial ischemia/reperfusion injury in transgenic mice. Biochim Biophys Acta 1773: 577–583, 2007. doi: 10.1016/j.bbamcr.2007.01.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Cogliati S, Frezza C, Soriano ME, Varanita T, Quintana-Cabrera R, Corrado M, Cipolat S, Costa V, Casarin A, Gomes LC, Perales-Clemente E, Salviati L, Fernandez-Silva P, Enriquez JA, Scorrano L. Mitochondrial cristae shape determines respiratory chain supercomplexes assembly and respiratory efficiency. Cell 155: 160–171, 2013. doi: 10.1016/j.cell.2013.08.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Cohen MV, Yang XM, Downey JM. The pH hypothesis of postconditioning: staccato reperfusion reintroduces oxygen and perpetuates myocardial acidosis. Circulation 115: 1895–1903, 2007. doi: 10.1161/CIRCULATIONAHA.106.675710. [DOI] [PubMed] [Google Scholar]
- 65.Communal C, Sumandea M, de Tombe P, Narula J, Solaro RJ, Hajjar RJ. Functional consequences of caspase activation in cardiac myocytes. Proc Natl Acad Sci USA 99: 6252–6256, 2002. doi: 10.1073/pnas.092022999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Connern CP, Halestrap AP. Purification and N-terminal sequencing of peptidyl-prolyl cis-trans-isomerase from rat liver mitochondrial matrix reveals the existence of a distinct mitochondrial cyclophilin. Biochem J 284: 381–385, 1992. doi: 10.1042/bj2840381. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Crompton M, Ellinger H, Costi A. Inhibition by cyclosporin A of a Ca2+-dependent pore in heart mitochondria activated by inorganic phosphate and oxidative stress. Biochem J 255: 357–360, 1988. [PMC free article] [PubMed] [Google Scholar]
- 68.Cuchet-Lourenço D, Eletto D, Wu C, Plagnol V, Papapietro O, Curtis J, Ceron-Gutierrez L, Bacon CM, Hackett S, Alsaleem B, Maes M, Gaspar M, Alisaac A, Goss E, AlIdrissi E, Siegmund D, Wajant H, Kumararatne D, AlZahrani MS, Arkwright PD, Abinun M, Doffinger R, Nejentsev S. Biallelic RIPK1 mutations in humans cause severe immunodeficiency, arthritis, and intestinal inflammation. Science 361: 810–813, 2018. doi: 10.1126/science.aar2641. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.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. [DOI] [PubMed] [Google Scholar]
- 70.Curtin NJ, Szabo C. Therapeutic applications of PARP inhibitors: anticancer therapy and beyond. Mol Aspects Med 34: 1217–1256, 2013. doi: 10.1016/j.mam.2013.01.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.D’Angelo DD, Sakata Y, Lorenz JN, Boivin GP, Walsh RA, Liggett SB, Dorn GW II. Transgenic Galphaq overexpression induces cardiac contractile failure in mice. Proc Natl Acad Sci USA 94: 8121–8126, 1997. doi: 10.1073/pnas.94.15.8121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Dabkowski ER, Williamson CL, Hollander JM. Mitochondria-specific transgenic overexpression of phospholipid hydroperoxide glutathione peroxidase (GPx4) attenuates ischemia/reperfusion-associated cardiac dysfunction. Free Radic Biol Med 45: 855–865, 2008. doi: 10.1016/j.freeradbiomed.2008.06.021. [DOI] [PubMed] [Google Scholar]
- 73.Danial NN, Korsmeyer SJ. Cell death: critical control points. Cell 116: 205–219, 2004. doi: 10.1016/S0092-8674(04)00046-7. [DOI] [PubMed] [Google Scholar]
- 74.Dannappel M, Vlantis K, Kumari S, Polykratis A, Kim C, Wachsmuth L, Eftychi C, Lin J, Corona T, Hermance N, Zelic M, Kirsch P, Basic M, Bleich A, Kelliher M, Pasparakis M. RIPK1 maintains epithelial homeostasis by inhibiting apoptosis and necroptosis. Nature 513: 90–94, 2014. doi: 10.1038/nature13608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Darmon AJ, Nicholson DW, Bleackley RC. Activation of the apoptotic protease CPP32 by cytotoxic T-cell-derived granzyme B. Nature 377: 446–448, 1995. doi: 10.1038/377446a0. [DOI] [PubMed] [Google Scholar]
- 76.Datta SR, Dudek H, Tao X, Masters S, Fu H, Gotoh Y, Greenberg ME. Akt phosphorylation of BAD couples survival signals to the cell-intrinsic death machinery. Cell 91: 231–241, 1997. doi: 10.1016/S0092-8674(00)80405-5. [DOI] [PubMed] [Google Scholar]
- 77.Daugas E, Nochy D, Ravagnan L, Loeffler M, Susin SA, Zamzami N, Kroemer G. Apoptosis-inducing factor (AIF): a ubiquitous mitochondrial oxidoreductase involved in apoptosis. FEBS Lett 476: 118–123, 2000. doi: 10.1016/S0014-5793(00)01731-2. [DOI] [PubMed] [Google Scholar]
- 78.David KK, Andrabi SA, Dawson TM, Dawson VL. Parthanatos, a messenger of death. Front Biosci 14: 1116–1128, 2009. doi: 10.2741/3297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.De Brito OM, Scorrano L. Mitofusin 2 tethers endoplasmic reticulum to mitochondria. [Corrigendum in Nature 513: 266, 2014.] Nature 456: 605–610, 2008. doi: 10.1038/nature07534. [DOI] [PubMed] [Google Scholar]
- 80.Decker RS, Poole AR, Crie JS, Dingle JT, Wildenthal K. Lysosomal alterations in hypoxic and reoxygenated hearts. II. Immunohistochemical and biochemical changes in cathepsin D. Am J Pathol 98: 445–456, 1980. [PMC free article] [PubMed] [Google Scholar]
- 81.Degterev A, Hitomi J, Germscheid M, Ch’en IL, Korkina O, Teng X, Abbott D, Cuny GD, Yuan C, Wagner G, Hedrick SM, Gerber SA, Lugovskoy A, Yuan J. Identification of RIP1 kinase as a specific cellular target of necrostatins. Nat Chem Biol 4: 313–321, 2008. doi: 10.1038/nchembio.83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Degterev A, Huang Z, Boyce M, Li Y, Jagtap P, Mizushima N, Cuny GD, Mitchison TJ, Moskowitz MA, Yuan J. Chemical inhibitor of nonapoptotic cell death with therapeutic potential for ischemic brain injury. Nat Chem Biol 1: 112–119, 2005. doi: 10.1038/nchembio711. [DOI] [PubMed] [Google Scholar]
- 83.Del Peso L, González-García M, Page C, Herrera R, Nuñez G. Interleukin-3-induced phosphorylation of BAD through the protein kinase Akt. Science 278: 687–689, 1997. doi: 10.1126/science.278.5338.687. [DOI] [PubMed] [Google Scholar]
- 84.Denecker G, Hoste E, Gilbert B, Hochepied T, Ovaere P, Lippens S, Van den Broecke C, Van Damme P, D’Herde K, Hachem JP, Borgonie G, Presland RB, Schoonjans L, Libert C, Vandekerckhove J, Gevaert K, Vandenabeele P, Declercq W. Caspase-14 protects against epidermal UVB photodamage and water loss. Nat Cell Biol 9: 666–674, 2007. doi: 10.1038/ncb1597. [DOI] [PubMed] [Google Scholar]
- 85.Denecker G, Ovaere P, Vandenabeele P, Declercq W. Caspase-14 reveals its secrets. J Cell Biol 180: 451–458, 2008. doi: 10.1083/jcb.200709098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Denton D, Xu T, Kumar S. Autophagy as a pro-death pathway. Immunol Cell Biol 93: 35–42, 2015. doi: 10.1038/icb.2014.85. [DOI] [PubMed] [Google Scholar]
- 87.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Deveraux QL, Takahashi R, Salvesen GS, Reed JC. X-linked IAP is a direct inhibitor of cell-death proteases. Nature 388: 300–304, 1997. doi: 10.1038/40901. [DOI] [PubMed] [Google Scholar]
- 90.Dhingra R, Margulets V, Chowdhury SR, Thliveris J, Jassal D, Fernyhough P, Dorn GW II, Kirshenbaum LA. Bnip3 mediates doxorubicin-induced cardiac myocyte necrosis and mortality through changes in mitochondrial signaling. Proc Natl Acad Sci USA 111: E5537–E5544, 2014. doi: 10.1073/pnas.1414665111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Dickens LS, Powley IR, Hughes MA, MacFarlane M. The ‟complexitiesˮ of life and death: death receptor signalling platforms. Exp Cell Res 318: 1269–1277, 2012. doi: 10.1016/j.yexcr.2012.04.005. [DOI] [PubMed] [Google Scholar]
- 92.Dillon CP, Oberst A, Weinlich R, Janke LJ, Kang TB, Ben-Moshe T, Mak TW, Wallach D, Green DR. Survival function of the FADD-CASPASE-8-cFLIP(L) complex. Cell Rep 1: 401–407, 2012. doi: 10.1016/j.celrep.2012.03.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Dillon CP, Weinlich R, Rodriguez DA, Cripps JG, Quarato G, Gurung P, Verbist KC, Brewer TL, Llambi F, Gong YN, Janke LJ, Kelliher MA, Kanneganti TD, Green DR. RIPK1 blocks early postnatal lethality mediated by caspase-8 and RIPK3. Cell 157: 1189–1202, 2014. doi: 10.1016/j.cell.2014.04.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Ding J, Wang K, Liu W, She Y, Sun Q, Shi J, Sun H, Wang DC, Shao F. Pore-forming activity and structural autoinhibition of the gasdermin family. [Erratum in Nature 540: 150, 2016.] Nature 535: 111–116, 2016. doi: 10.1038/nature18590. [DOI] [PubMed] [Google Scholar]
- 95.Dispersyn GD, Ausma J, Thoné F, Flameng W, Vanoverschelde JL, Allessie MA, Ramaekers FC, Borgers M. Cardiomyocyte remodelling during myocardial hibernation and atrial fibrillation: prelude to apoptosis. Cardiovasc Res 43: 947–957, 1999. doi: 10.1016/S0008-6363(99)00096-6. [DOI] [PubMed] [Google Scholar]
- 96.Diwan A, Krenz M, Syed FM, Wansapura J, Ren X, Koesters AG, Li H, Kirshenbaum LA, Hahn HS, Robbins J, Jones WK, Dorn GW II. Inhibition of ischemic cardiomyocyte apoptosis through targeted ablation of Bnip3 restrains postinfarction remodeling in mice. J Clin Invest 117: 2825–2833, 2007. doi: 10.1172/JCI32490. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, Patel DN, Bauer AJ, Cantley AM, Yang WS, Morrison B III, Stockwell BR. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell 149: 1060–1072, 2012. doi: 10.1016/j.cell.2012.03.042. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Docherty JC, Kuzio B, Silvester JA, Bowes J, Thiemermann C. An inhibitor of poly (ADP-ribose) synthetase activity reduces contractile dysfunction and preserves high energy phosphate levels during reperfusion of the ischaemic rat heart. Br J Pharmacol 127: 1518–1524, 1999. doi: 10.1038/sj.bjp.0702705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Donath S, Li P, Willenbockel C, Al-Saadi N, Gross V, Willnow T, Bader M, Martin U, Bauersachs J, Wollert KC, Dietz R, von Harsdorf R; German Heart Failure Network . Apoptosis repressor with caspase recruitment domain is required for cardioprotection in response to biomechanical and ischemic stress. Circulation 113: 1203–1212, 2006. doi: 10.1161/CIRCULATIONAHA.105.576785. [DOI] [PubMed] [Google Scholar]
- 100.Dondelinger Y, Declercq W, Montessuit S, Roelandt R, Goncalves A, Bruggeman I, Hulpiau P, Weber K, Sehon CA, Marquis RW, Bertin J, Gough PJ, Savvides S, Martinou JC, Bertrand MJ, Vandenabeele P. MLKL compromises plasma membrane integrity by binding to phosphatidylinositol phosphates. Cell Rep 7: 971–981, 2014. doi: 10.1016/j.celrep.2014.04.026. [DOI] [PubMed] [Google Scholar]
- 101.Dondelinger Y, Delanghe T, Rojas-Rivera D, Priem D, Delvaeye T, Bruggeman I, Van Herreweghe F, Vandenabeele P, Bertrand MJM. MK2 phosphorylation of RIPK1 regulates TNF-mediated cell death. Nat Cell Biol 19: 1237–1247, 2017. doi: 10.1038/ncb3608. [DOI] [PubMed] [Google Scholar]
- 102.Dondelinger Y, Jouan-Lanhouet S, Divert T, Theatre E, Bertin J, Gough PJ, Giansanti P, Heck AJ, Dejardin E, Vandenabeele P, Bertrand MJ. NF-κB-Independent Role of IKKα/IKKβ in Preventing RIPK1 Kinase-Dependent Apoptotic and Necroptotic Cell Death during TNF Signaling. Mol Cell 60: 63–76, 2015. doi: 10.1016/j.molcel.2015.07.032. [DOI] [PubMed] [Google Scholar]
- 103.Dong H, Zhu G, Tamada K, Chen L. B7-H1, a third member of the B7 family, co-stimulates T-cell proliferation and interleukin-10 secretion. Nat Med 5: 1365–1369, 1999. doi: 10.1038/70932. [DOI] [PubMed] [Google Scholar]
- 104.Dovey CM, Diep J, Clarke BP, Hale AT, McNamara DE, Guo H, Brown NW Jr, Cao JY, Grace CR, Gough PJ, Bertin J, Dixon SJ, Fiedler D, Mocarski ES, Kaiser WJ, Moldoveanu T, York JD, Carette JE. MLKL Requires the Inositol Phosphate Code to Execute Necroptosis. Mol Cell 70: 936–948.e7, 2018. doi: 10.1016/j.molcel.2018.05.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Draber P, Kupka S, Reichert M, Draberova H, Lafont E, de Miguel D, Spilgies L, Surinova S, Taraborrelli L, Hartwig T, Rieser E, Martino L, Rittinger K, Walczak H. LUBAC-Recruited CYLD and A20 Regulate Gene Activation and Cell Death by Exerting Opposing Effects on Linear Ubiquitin in Signaling Complexes. Cell Rep 13: 2258–2272, 2015. doi: 10.1016/j.celrep.2015.11.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Du C, Fang M, Li Y, Li L, Wang X. Smac, a mitochondrial protein that promotes cytochrome c-dependent caspase activation by eliminating IAP inhibition. Cell 102: 33–42, 2000. doi: 10.1016/S0092-8674(00)00008-8. [DOI] [PubMed] [Google Scholar]
- 107.Du S, Zhou L, Alexander GS, Park K, Yang L, Wang N, Zaorsky NG, Ma X, Wang Y, Dicker AP, Lu B. PD-1 Modulates Radiation-Induced Cardiac Toxicity through Cytotoxic T Lymphocytes. J Thorac Oncol 13: 510–520, 2018. doi: 10.1016/j.jtho.2017.12.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Duckett CS, Nava VE, Gedrich RW, Clem RJ, Van Dongen JL, Gilfillan MC, Shiels H, Hardwick JM, Thompson CB. A conserved family of cellular genes related to the baculovirus iap gene and encoding apoptosis inhibitors. EMBO J 15: 2685–2694, 1996. doi: 10.1002/j.1460-2075.1996.tb00629.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Dziedzic SA, Su Z, Jean Barrett V, Najafov A, Mookhtiar AK, Amin P, Pan H, Sun L, Zhu H, Ma A, Abbott DW, Yuan J. ABIN-1 regulates RIPK1 activation by linking Met1 ubiquitylation with Lys63 deubiquitylation in TNF-RSC. Nat Cell Biol 20: 58–68, 2018. doi: 10.1038/s41556-017-0003-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Ea CK, Deng L, Xia ZP, Pineda G, Chen ZJ. Activation of IKK by TNFalpha requires site-specific ubiquitination of RIP1 and polyubiquitin binding by NEMO. Mol Cell 22: 245–257, 2006. doi: 10.1016/j.molcel.2006.03.026. [DOI] [PubMed] [Google Scholar]
- 111.Edlich F, Banerjee S, Suzuki M, Cleland MM, Arnoult D, Wang C, Neutzner A, Tjandra N, Youle RJ. Bcl-x(L) retrotranslocates Bax from the mitochondria into the cytosol. Cell 145: 104–116, 2011. doi: 10.1016/j.cell.2011.02.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Ekert PG, Vaux DL. The mitochondrial death squad: hardened killers or innocent bystanders? Curr Opin Cell Biol 17: 626–630, 2005. doi: 10.1016/j.ceb.2005.09.001. [DOI] [PubMed] [Google Scholar]
- 113.Elliott PR, Leske D, Hrdinka M, Bagola K, Fiil BK, McLaughlin SH, Wagstaff J, Volkmar N, Christianson JC, Kessler BM, Freund SM, Komander D, Gyrd-Hansen M. SPATA2 Links CYLD to LUBAC, Activates CYLD, and Controls LUBAC Signaling. Mol Cell 63: 990–1005, 2016. doi: 10.1016/j.molcel.2016.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Elrod JW, Wong R, Mishra S, Vagnozzi RJ, Sakthievel B, Goonasekera SA, Karch J, Gabel S, Farber J, Force T, Brown JH, Murphy E, Molkentin JD. Cyclophilin D controls mitochondrial pore-dependent Ca2+ exchange, metabolic flexibility, and propensity for heart failure in mice. J Clin Invest 120: 3680–3687, 2010. doi: 10.1172/JCI43171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Enari M, Sakahira H, Yokoyama H, Okawa K, Iwamatsu A, Nagata S. A caspase-activated DNase that degrades DNA during apoptosis, and its inhibitor ICAD. Nature 391: 43–50, 1998. doi: 10.1038/34112. [DOI] [PubMed] [Google Scholar]
- 116.Eppihimer MJ, Gunn J, Freeman GJ, Greenfield EA, Chernova T, Erickson J, Leonard JP. Expression and regulation of the PD-L1 immunoinhibitory molecule on microvascular endothelial cells. Microcirculation 9: 133–145, 2002. doi: 10.1080/713774061. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Esmann M, Fedosova NU, Marsh D. Osmotic stress and viscous retardation of the Na,K-ATPase ion pump. Biophys J 94: 2767–2776, 2008. doi: 10.1529/biophysj.106.101774. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Faccio L, Fusco C, Chen A, Martinotti S, Bonventre JV, Zervos AS. Characterization of a novel human serine protease that has extensive homology to bacterial heat shock endoprotease HtrA and is regulated by kidney ischemia. J Biol Chem 275: 2581–2588, 2000. doi: 10.1074/jbc.275.4.2581. [DOI] [PubMed] [Google Scholar]
- 119.Fan Y, Yu Y, Shi Y, Sun W, Xie M, Ge N, Mao R, Chang A, Xu G, Schneider MD, Zhang H, Fu S, Qin J, Yang J. Lysine 63-linked polyubiquitination of TAK1 at lysine 158 is required for tumor necrosis factor alpha- and interleukin-1beta-induced IKK/NF-kappaB and JNK/AP-1 activation. J Biol Chem 285: 5347–5360, 2010. doi: 10.1074/jbc.M109.076976. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Fang X, Wang H, Han D, Xie E, Yang X, Wei J, Gu S, Gao F, Zhu N, Yin X, Cheng Q, Zhang P, Dai W, Chen J, Yang F, Yang HT, Linkermann A, Gu W, Min J, Wang F. Ferroptosis as a target for protection against cardiomyopathy. Proc Natl Acad Sci USA 116: 2672–2680, 2019. doi: 10.1073/pnas.1821022116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Fatokun AA, Dawson VL, Dawson TM. Parthanatos: mitochondrial-linked mechanisms and therapeutic opportunities. Br J Pharmacol 171: 2000–2016, 2014. doi: 10.1111/bph.12416. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Faustman D, Davis M. TNF receptor 2 pathway: drug target for autoimmune diseases. Nat Rev Drug Discov 9: 482–493, 2010. doi: 10.1038/nrd3030. [DOI] [PubMed] [Google Scholar]
- 123.Feng H, Stockwell BR. Unsolved mysteries: how does lipid peroxidation cause ferroptosis? PLoS Biol 16: e2006203, 2018. doi: 10.1371/journal.pbio.2006203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Feng S, Yang Y, Mei Y, Ma L, Zhu DE, Hoti N, Castanares M, Wu M. Cleavage of RIP3 inactivates its caspase-independent apoptosis pathway by removal of kinase domain. Cell Signal 19: 2056–2067, 2007. doi: 10.1016/j.cellsig.2007.05.016. [DOI] [PubMed] [Google Scholar]
- 125.Feoktistova M, Geserick P, Kellert B, Dimitrova DP, Langlais C, Hupe M, Cain K, MacFarlane M, Häcker G, Leverkus M. cIAPs block Ripoptosome formation, a RIP1/caspase-8 containing intracellular cell death complex differentially regulated by cFLIP isoforms. Mol Cell 43: 449–463, 2011. doi: 10.1016/j.molcel.2011.06.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Fernandes-Alnemri T, Kang S, Anderson C, Sagara J, Fitzgerald KA, Alnemri ES. Cutting edge: TLR signaling licenses IRAK1 for rapid activation of the NLRP3 inflammasome. J Immunol 191: 3995–3999, 2013. doi: 10.4049/jimmunol.1301681. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Fillmore N, Mori J, Lopaschuk GD. Mitochondrial fatty acid oxidation alterations in heart failure, ischaemic heart disease and diabetic cardiomyopathy. Br J Pharmacol 171: 2080–2090, 2014. doi: 10.1111/bph.12475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Fink SL, Cookson BT. Caspase-1-dependent pore formation during pyroptosis leads to osmotic lysis of infected host macrophages. Cell Microbiol 8: 1812–1825, 2006. doi: 10.1111/j.1462-5822.2006.00751.x. [DOI] [PubMed] [Google Scholar]
- 129.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Foo RS, Chan LK, Kitsis RN, Bennett MR. Ubiquitination and degradation of the anti-apoptotic protein ARC by MDM2. J Biol Chem 282: 5529–5535, 2007. doi: 10.1074/jbc.M609046200. [DOI] [PubMed] [Google Scholar]
- 131.Foo RS, Nam YJ, Ostreicher MJ, Metzl MD, Whelan RS, Peng CF, Ashton AW, Fu W, Mani K, Chin SF, Provenzano E, Ellis I, Figg N, Pinder S, Bennett MR, Caldas C, Kitsis RN. Regulation of p53 tetramerization and nuclear export by ARC. Proc Natl Acad Sci USA 104: 20826–20831, 2007. doi: 10.1073/pnas.0710017104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Frank S, Gaume B, Bergmann-Leitner ES, Leitner WW, Robert EG, Catez F, Smith CL, Youle RJ. The role of dynamin-related protein 1, a mediator of mitochondrial fission, in apoptosis. Dev Cell 1: 515–525, 2001. doi: 10.1016/S1534-5807(01)00055-7. [DOI] [PubMed] [Google Scholar]
- 133.Freeman GJ, Long AJ, Iwai Y, Bourque K, Chernova T, Nishimura H, Fitz LJ, Malenkovich N, Okazaki T, Byrne MC, Horton HF, Fouser L, Carter L, Ling V, Bowman MR, Carreno BM, Collins M, Wood CR, Honjo T. Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J Exp Med 192: 1027–1034, 2000. doi: 10.1084/jem.192.7.1027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Friedmann Angeli JP, Schneider M, Proneth B, Tyurina YY, Tyurin VA, Hammond VJ, Herbach N, Aichler M, Walch A, Eggenhofer E, Basavarajappa D, Rådmark O, Kobayashi S, Seibt T, Beck H, Neff F, Esposito I, Wanke R, Förster H, Yefremova O, Heinrichmeyer M, Bornkamm GW, Geissler EK, Thomas SB, Stockwell BR, O’Donnell VB, Kagan VE, Schick JA, Conrad M. Inactivation of the ferroptosis regulator Gpx4 triggers acute renal failure in mice. Nat Cell Biol 16: 1180–1191, 2014. doi: 10.1038/ncb3064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Fu Q, Fu TM, Cruz AC, Sengupta P, Thomas SK, Wang S, Siegel RM, Wu H, Chou JJ. Structural Basis and Functional Role of Intramembrane Trimerization of the Fas/CD95 Death Receptor. Mol Cell 61: 602–613, 2016. doi: 10.1016/j.molcel.2016.01.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Fuchs TA, Abed U, Goosmann C, Hurwitz R, Schulze I, Wahn V, Weinrauch Y, Brinkmann V, Zychlinsky A. Novel cell death program leads to neutrophil extracellular traps. J Cell Biol 176: 231–241, 2007. doi: 10.1083/jcb.200606027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Fujio Y, Nguyen T, Wencker D, Kitsis RN, Walsh K. Akt promotes survival of cardiomyocytes in vitro and protects against ischemia-reperfusion injury in mouse heart. Circulation 101: 660–667, 2000. doi: 10.1161/01.CIR.101.6.660. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Fujita H, Tokunaga A, Shimizu S, Whiting AL, Aguilar-Alonso F, Takagi K, Walinda E, Sasaki Y, Shimokawa T, Mizushima T, Ohki I, Ariyoshi M, Tochio H, Bernal F, Shirakawa M, Iwai K. Cooperative Domain Formation by Homologous Motifs in HOIL-1L and SHARPIN Plays a Crucial Role in LUBAC Stabilization. Cell Rep 23: 1192–1204, 2018. doi: 10.1016/j.celrep.2018.03.112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Galluzzi L, Baehrecke EH, Ballabio A, Boya P, Bravo-San Pedro JM, Cecconi F, Choi AM, Chu CT, Codogno P, Colombo MI, Cuervo AM, Debnath J, Deretic V, Dikic I, Eskelinen EL, Fimia GM, Fulda S, Gewirtz DA, Green DR, Hansen M, Harper JW, Jäättelä M, Johansen T, Juhasz G, Kimmelman AC, Kraft C, Ktistakis NT, Kumar S, Levine B, Lopez-Otin C, Madeo F, Martens S, Martinez J, Melendez A, Mizushima N, Münz C, Murphy LO, Penninger JM, Piacentini M, Reggiori F, Rubinsztein DC, Ryan KM, Santambrogio L, Scorrano L, Simon AK, Simon HU, Simonsen A, Tavernarakis N, Tooze SA, Yoshimori T, Yuan J, Yue Z, Zhong Q, Kroemer G. Molecular definitions of autophagy and related processes. EMBO J 36: 1811–1836, 2017. doi: 10.15252/embj.201796697. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Galluzzi L, Buqué A, Kepp O, Zitvogel L, Kroemer G. Immunogenic cell death in cancer and infectious disease. Nat Rev Immunol 17: 97–111, 2017. doi: 10.1038/nri.2016.107. [DOI] [PubMed] [Google Scholar]
- 141.Galluzzi L, Chan TA, Kroemer G, Wolchok JD, López-Soto A. The hallmarks of successful anticancer immunotherapy. Sci Transl Med 10: eaat7807, 2018. doi: 10.1126/scitranslmed.aat7807. [DOI] [PubMed] [Google Scholar]
- 142.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Galluzzi L, Yamazaki T, Kroemer G. Linking cellular stress responses to systemic homeostasis. Nat Rev Mol Cell Biol 19: 731–745, 2018. doi: 10.1038/s41580-018-0068-0. [DOI] [PubMed] [Google Scholar]
- 144.Gao M, Monian P, Quadri N, Ramasamy R, Jiang X. Glutaminolysis and Transferrin Regulate Ferroptosis. Mol Cell 59: 298–308, 2015. doi: 10.1016/j.molcel.2015.06.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Garcia-Diaz A, Shin DS, Moreno BH, Saco J, Escuin-Ordinas H, Rodriguez GA, Zaretsky JM, Sun L, Hugo W, Wang X, Parisi G, Saus CP, Torrejon DY, Graeber TG, Comin-Anduix B, Hu-Lieskovan S, Damoiseaux R, Lo RS, Ribas A. Interferon Receptor Signaling Pathways Regulating PD-L1 and PD-L2 Expression. Cell Rep 19: 1189–1201, 2017. doi: 10.1016/j.celrep.2017.04.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Garner TP, Amgalan D, Reyna DE, Li S, Kitsis RN, Gavathiotis E. Small-molecule allosteric inhibitors of BAX. Nat Chem Biol 15: 322–330, 2019. doi: 10.1038/s41589-018-0223-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Gavathiotis E, Reyna DE, Davis ML, Bird GH, Walensky LD. BH3-triggered structural reorganization drives the activation of proapoptotic BAX. Mol Cell 40: 481–492, 2010. doi: 10.1016/j.molcel.2010.10.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Gavathiotis E, Suzuki M, Davis ML, Pitter K, Bird GH, Katz SG, Tu HC, Kim H, Cheng EH, Tjandra N, Walensky LD. BAX activation is initiated at a novel interaction site. Nature 455: 1076–1081, 2008. doi: 10.1038/nature07396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Geertman R, McMahon A, Sabban EL. Cloning and characterization of cDNAs for novel proteins with glutamic acid-proline dipeptide tandem repeats. Biochim Biophys Acta 1306: 147–152, 1996. doi: 10.1016/0167-4781(96)00036-X. [DOI] [PubMed] [Google Scholar]
- 150.Geng J, Ito Y, Shi L, Amin P, Chu J, Ouchida AT, Mookhtiar AK, Zhao H, Xu D, Shan B, Najafov A, Gao G, Akira S, Yuan J. Regulation of RIPK1 activation by TAK1-mediated phosphorylation dictates apoptosis and necroptosis. Nat Commun 8: 359, 2017. doi: 10.1038/s41467-017-00406-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.George NM, Evans JJ, Luo X. A three-helix homo-oligomerization domain containing BH3 and BH1 is responsible for the apoptotic activity of Bax. Genes Dev 21: 1937–1948, 2007. doi: 10.1101/gad.1553607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Glücksmann A. Cell deaths in normal vertebrate ontogeny. Biol Rev Camb Philos Soc 26: 59–86, 1951. doi: 10.1111/j.1469-185X.1951.tb00774.x. [DOI] [PubMed] [Google Scholar]
- 154.Gong YN, Guy C, Olauson H, Becker JU, Yang M, Fitzgerald P, Linkermann A, Green DR. ESCRT-III Acts Downstream of MLKL to Regulate Necroptotic Cell Death and Its Consequences. Cell 169: 286–300.e16, 2017. doi: 10.1016/j.cell.2017.03.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Goping IS, Barry M, Liston P, Sawchuk T, Constantinescu G, Michalak KM, Shostak I, Roberts DL, Hunter AM, Korneluk R, Bleackley RC. Granzyme B-induced apoptosis requires both direct caspase activation and relief of caspase inhibition. Immunity 18: 355–365, 2003. doi: 10.1016/S1074-7613(03)00032-3. [DOI] [PubMed] [Google Scholar]
- 156.Gottlieb RA, Burleson KO, Kloner RA, Babior BM, Engler RL. Reperfusion injury induces apoptosis in rabbit cardiomyocytes. J Clin Invest 94: 1621–1628, 1994. doi: 10.1172/JCI117504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Grabie N, Gotsman I, DaCosta R, Pang H, Stavrakis G, Butte MJ, Keir ME, Freeman GJ, Sharpe AH, Lichtman AH. Endothelial programmed death-1 ligand 1 (PD-L1) regulates CD8+ T-cell mediated injury in the heart. Circulation 116: 2062–2071, 2007. doi: 10.1161/CIRCULATIONAHA.107.709360. [DOI] [PubMed] [Google Scholar]
- 158.Grabie N, Lichtman AH, Padera R. T cell checkpoint regulators in the heart. Cardiovasc Res 115: 869–877, 2019. doi: 10.1093/cvr/cvz025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Green DR, Ferguson TA. The role of Fas ligand in immune privilege. Nat Rev Mol Cell Biol 2: 917–924, 2001. doi: 10.1038/35103104. [DOI] [PubMed] [Google Scholar]
- 160.Griendling KK, Sorescu D, Ushio-Fukai M. NAD(P)H oxidase: role in cardiovascular biology and disease. Circ Res 86: 494–501, 2000. doi: 10.1161/01.RES.86.5.494. [DOI] [PubMed] [Google Scholar]
- 161.Griffith TS, Brunner T, Fletcher SM, Green DR, Ferguson TA. Fas ligand-induced apoptosis as a mechanism of immune privilege. Science 270: 1189–1192, 1995. doi: 10.1126/science.270.5239.1189. [DOI] [PubMed] [Google Scholar]
- 162.Griffiths EJ, Ocampo CJ, Savage JS, Rutter GA, Hansford RG, Stern MD, Silverman HS. Mitochondrial calcium transporting pathways during hypoxia and reoxygenation in single rat cardiomyocytes. Cardiovasc Res 39: 423–433, 1998. doi: 10.1016/S0008-6363(98)00104-7. [DOI] [PubMed] [Google Scholar]
- 163.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Gross A, Yin XM, Wang K, Wei MC, Jockel J, Milliman C, Erdjument-Bromage H, Tempst P, Korsmeyer SJ. Caspase cleaved BID targets mitochondria and is required for cytochrome c release, while BCL-XL prevents this release but not tumor necrosis factor-R1/Fas death. J Biol Chem 274: 1156–1163, 1999. doi: 10.1074/jbc.274.2.1156. [DOI] [PubMed] [Google Scholar]
- 165.Grupp IL, Jackson TM, Hake P, Grupp G, Szabó C. Protection against hypoxia-reoxygenation in the absence of poly (ADP-ribose) synthetase in isolated working hearts. J Mol Cell Cardiol 31: 297–303, 1999. doi: 10.1006/jmcc.1998.0864. [DOI] [PubMed] [Google Scholar]
- 166.Guan P, Wang N. Mammalian target of rapamycin coordinates iron metabolism with iron-sulfur cluster assembly enzyme and tristetraprolin. Nutrition 30: 968–974, 2014. doi: 10.1016/j.nut.2013.12.016. [DOI] [PubMed] [Google Scholar]
- 167.Guerra S, Leri A, Wang X, Finato N, Di Loreto C, Beltrami CA, Kajstura J, Anversa P. Myocyte death in the failing human heart is gender dependent. Circ Res 85: 856–866, 1999. doi: 10.1161/01.RES.85.9.856. [DOI] [PubMed] [Google Scholar]
- 168.Guo K, Searfoss G, Krolikowski D, Pagnoni M, Franks C, Clark K, Yu KT, Jaye M, Ivashchenko Y. Hypoxia induces the expression of the pro-apoptotic gene BNIP3. Cell Death Differ 8: 367–376, 2001. doi: 10.1038/sj.cdd.4400810. [DOI] [PubMed] [Google Scholar]
- 169.Guo X, Yin H, Li L, Chen Y, Li J, Doan J, Steinmetz RN, Liu Q. Cardioprotective Role of Tumor Necrosis Factor Receptor-Associated Factor 2 by Suppressing Apoptosis and Necroptosis. Circulation 136: 729–742, 2017. doi: 10.1161/CIRCULATIONAHA.116.026240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Gurung P, Anand PK, Malireddi RK, Vande Walle L, Van Opdenbosch N, Dillon CP, Weinlich R, Green DR, Lamkanfi M, Kanneganti TD. FADD and caspase-8 mediate priming and activation of the canonical and noncanonical Nlrp3 inflammasomes. J Immunol 192: 1835–1846, 2014. doi: 10.4049/jimmunol.1302839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Gutiérrez-Aguilar M, Douglas DL, Gibson AK, Domeier TL, Molkentin JD, Baines CP. Genetic manipulation of the cardiac mitochondrial phosphate carrier does not affect permeability transition. J Mol Cell Cardiol 72: 316–325, 2014. doi: 10.1016/j.yjmcc.2014.04.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Halestrap AP, Davidson AM. Inhibition of Ca2+-induced large-amplitude swelling of liver and heart mitochondria by cyclosporin is probably caused by the inhibitor binding to mitochondrial-matrix peptidyl-prolyl cis-trans isomerase and preventing it interacting with the adenine nucleotide translocase. Biochem J 268: 153–160, 1990. doi: 10.1042/bj2680153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Halestrap AP, Richardson AP. The mitochondrial permeability transition: a current perspective on its identity and role in ischaemia/reperfusion injury. J Mol Cell Cardiol 78: 129–141, 2015. doi: 10.1016/j.yjmcc.2014.08.018. [DOI] [PubMed] [Google Scholar]
- 174.Hall AR, Burke N, Dongworth RK, Kalkhoran SB, Dyson A, Vicencio JM, Dorn GW II, Yellon DM, Hausenloy DJ. Hearts deficient in both Mfn1 and Mfn2 are protected against acute myocardial infarction. Cell Death Dis 7: e2238, 2016. doi: 10.1038/cddis.2016.139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Hamburger V, Levi-Montalcini R. Proliferation, differentiation and degeneration in the spinal ganglia of the chick embryo under normal and experimental conditions. J Exp Zool 111: 457–501, 1949. doi: 10.1002/jez.1401110308. [DOI] [PubMed] [Google Scholar]
- 176.Han S, Lear TB, Jerome JA, Rajbhandari S, Snavely CA, Gulick DL, Gibson KF, Zou C, Chen BB, Mallampalli RK. Lipopolysaccharide Primes the NALP3 Inflammasome by Inhibiting Its Ubiquitination and Degradation Mediated by the SCFFBXL2 E3 Ligase. J Biol Chem 290: 18124–18133, 2015. doi: 10.1074/jbc.M115.645549. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Harris JL, Peterson EP, Hudig D, Thornberry NA, Craik CS. Definition and redesign of the extended substrate specificity of granzyme B. J Biol Chem 273: 27364–27373, 1998. doi: 10.1074/jbc.273.42.27364. [DOI] [PubMed] [Google Scholar]
- 178.Hausenloy DJ, Yellon DM. Myocardial ischemia-reperfusion injury: a neglected therapeutic target. J Clin Invest 123: 92–100, 2013. doi: 10.1172/JCI62874. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Haworth RA, Hunter DR. Control of the mitochondrial permeability transition pore by high-affinity ADP binding at the ADP/ATP translocase in permeabilized mitochondria. J Bioenerg Biomembr 32: 91–96, 2000. doi: 10.1023/A:1005568630151. [DOI] [PubMed] [Google Scholar]
- 180.Hayakawa Y, Chandra M, Miao W, Shirani J, Brown JH, Dorn GW II, Armstrong RC, Kitsis RN. Inhibition of cardiac myocyte apoptosis improves cardiac function and abolishes mortality in the peripartum cardiomyopathy of Galpha(q) transgenic mice. Circulation 108: 3036–3041, 2003. doi: 10.1161/01.CIR.0000101920.72665.58. [DOI] [PubMed] [Google Scholar]
- 181.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.He S, Wang L, Miao L, Wang T, Du F, Zhao L, Wang X. Receptor interacting protein kinase-3 determines cellular necrotic response to TNF-alpha. Cell 137: 1100–1111, 2009. doi: 10.1016/j.cell.2009.05.021. [DOI] [PubMed] [Google Scholar]
- 184.He WT, Wan H, Hu L, Chen P, Wang X, Huang Z, Yang ZH, Zhong CQ, Han J. Gasdermin D is an executor of pyroptosis and required for interleukin-1β secretion. Cell Res 25: 1285–1298, 2015. doi: 10.1038/cr.2015.139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Hegde R, Srinivasula SM, Zhang Z, Wassell R, Mukattash R, Cilenti L, DuBois G, Lazebnik Y, Zervos AS, Fernandes-Alnemri T, Alnemri ES. Identification of Omi/HtrA2 as a mitochondrial apoptotic serine protease that disrupts inhibitor of apoptosis protein-caspase interaction. J Biol Chem 277: 432–438, 2002. doi: 10.1074/jbc.M109721200. [DOI] [PubMed] [Google Scholar]
- 186.Heibein JA, Goping IS, Barry M, Pinkoski MJ, Shore GC, Green DR, Bleackley RC. Granzyme B-mediated cytochrome c release is regulated by the Bcl-2 family members bid and Bax. J Exp Med 192: 1391–1402, 2000. doi: 10.1084/jem.192.10.1391. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Hengartner MO, Horvitz HR. C. elegans cell survival gene ced-9 encodes a functional homolog of the mammalian proto-oncogene bcl-2. Cell 76: 665–676, 1994. doi: 10.1016/0092-8674(94)90506-1. [DOI] [PubMed] [Google Scholar]
- 188.Hernandez-Cuellar E, Tsuchiya K, Hara H, Fang R, Sakai S, Kawamura I, Akira S, Mitsuyama M. Cutting edge: nitric oxide inhibits the NLRP3 inflammasome. J Immunol 189: 5113–5117, 2012. doi: 10.4049/jimmunol.1202479. [DOI] [PubMed] [Google Scholar]
- 189.Hildebrand JM, Tanzer MC, Lucet IS, Young SN, Spall SK, Sharma P, Pierotti C, Garnier JM, Dobson RC, Webb AI, Tripaydonis A, Babon JJ, Mulcair MD, Scanlon MJ, Alexander WS, Wilks AF, Czabotar PE, Lessene G, Murphy JM, Silke J. Activation of the pseudokinase MLKL unleashes the four-helix bundle domain to induce membrane localization and necroptotic cell death. Proc Natl Acad Sci USA 111: 15072–15077, 2014. doi: 10.1073/pnas.1408987111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Hitomi J, Christofferson DE, Ng A, Yao J, Degterev A, Xavier RJ, Yuan J. Identification of a molecular signaling network that regulates a cellular necrotic cell death pathway. Cell 135: 1311–1323, 2008. doi: 10.1016/j.cell.2008.10.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.Hochhauser E, Cheporko Y, Yasovich N, Pinchas L, Offen D, Barhum Y, Pannet H, Tobar A, Vidne BA, Birk E. Bax deficiency reduces infarct size and improves long-term function after myocardial infarction. Cell Biochem Biophys 47: 11–19, 2007. doi: 10.1385/CBB:47:1:11. [DOI] [PubMed] [Google Scholar]
- 192.Hochhauser E, Kivity S, Offen D, Maulik N, Otani H, Barhum Y, Pannet H, Shneyvays V, Shainberg A, Goldshtaub V, Tobar A, Vidne BA. Bax ablation protects against myocardial ischemia-reperfusion injury in transgenic mice. Am J Physiol Heart Circ Physiol 284: H2351–H2359, 2003. doi: 10.1152/ajpheart.00783.2002. [DOI] [PubMed] [Google Scholar]
- 193.Hofer-Warbinek R, Schmid JA, Stehlik C, Binder BR, Lipp J, de Martin R. Activation of NF-kappa B by XIAP, the X chromosome-linked inhibitor of apoptosis, in endothelial cells involves TAK1. J Biol Chem 275: 22064–22068, 2000. doi: 10.1074/jbc.M910346199. [DOI] [PubMed] [Google Scholar]
- 194.Holler N, Zaru R, Micheau O, Thome M, Attinger A, Valitutti S, Bodmer JL, Schneider P, Seed B, Tschopp J. Fas triggers an alternative, caspase-8-independent cell death pathway using the kinase RIP as effector molecule. Nat Immunol 1: 489–495, 2000. doi: 10.1038/82732. [DOI] [PubMed] [Google Scholar]
- 195.Holly TA, Drincic A, Byun Y, Nakamura S, Harris K, Klocke FJ, Cryns VL. Caspase inhibition reduces myocyte cell death induced by myocardial ischemia and reperfusion in vivo. J Mol Cell Cardiol 31: 1709–1715, 1999. doi: 10.1006/jmcc.1999.1006. [DOI] [PubMed] [Google Scholar]
- 196.Hoppins S, Edlich F, Cleland MM, Banerjee S, McCaffery JM, Youle RJ, Nunnari J. The soluble form of Bax regulates mitochondrial fusion via MFN2 homotypic complexes. Mol Cell 41: 150–160, 2011. doi: 10.1016/j.molcel.2010.11.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Hu JR, Florido R, Lipson EJ, Naidoo J, Ardehali R, Tocchetti CG, Lyon AR, Padera RF, Johnson DB, Moslehi J. Cardiovascular toxicities associated with immune checkpoint inhibitors. Cardiovasc Res 115: 854–868, 2019. doi: 10.1093/cvr/cvz026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Huang JQ, Radinovic S, Rezaiefar P, Black SC. In vivo myocardial infarct size reduction by a caspase inhibitor administered after the onset of ischemia. Eur J Pharmacol 402: 139–142, 2000. doi: 10.1016/S0014-2999(00)00477-5. [DOI] [PubMed] [Google Scholar]
- 199.Huang Y, Park YC, Rich RL, Segal D, Myszka DG, Wu H. Structural basis of caspase inhibition by XIAP: differential roles of the linker versus the BIR domain. Cell 104: 781–790, 2001. doi: 10.1016/S0092-8674(02)02075-5. [DOI] [PubMed] [Google Scholar]
- 200.Hughes MA, Powley IR, Jukes-Jones R, Horn S, Feoktistova M, Fairall L, Schwabe JW, Leverkus M, Cain K, MacFarlane M. Co-operative and Hierarchical Binding of c-FLIP and Caspase-8: A Unified Model Defines How c-FLIP Isoforms Differentially Control Cell Fate. Mol Cell 61: 834–849, 2016. doi: 10.1016/j.molcel.2016.02.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Hui E, Cheung J, Zhu J, Su X, Taylor MJ, Wallweber HA, Sasmal DK, Huang J, Kim JM, Mellman I, Vale RD. T cell costimulatory receptor CD28 is a primary target for PD-1-mediated inhibition. Science 355: 1428–1433, 2017. doi: 10.1126/science.aaf1292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Humphries F, Bergin R, Jackson R, Delagic N, Wang B, Yang S, Dubois AV, Ingram RJ, Moynagh PN. The E3 ubiquitin ligase Pellino2 mediates priming of the NLRP3 inflammasome. Nat Commun 9: 1560, 2018. doi: 10.1038/s41467-018-03669-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Hunter DR, Haworth RA. The Ca2+-induced membrane transition in mitochondria. I. The protective mechanisms. Arch Biochem Biophys 195: 453–459, 1979. doi: 10.1016/0003-9861(79)90371-0. [DOI] [PubMed] [Google Scholar]
- 204.Hunter FE Jr, Ford L. Inactivation of oxidative and phosphorylative systems in mitochondria by preincubation with phosphate and other ions. J Biol Chem 216: 357–369, 1955. [PubMed] [Google Scholar]
- 205.Imahashi K, Pott C, Goldhaber JI, Steenbergen C, Philipson KD, Murphy E. Cardiac-specific ablation of the Na+-Ca2+ exchanger confers protection against ischemia/reperfusion injury. Circ Res 97: 916–921, 2005. doi: 10.1161/01.RES.0000187456.06162.cb. [DOI] [PubMed] [Google Scholar]
- 206.Inserte J, Cardona M, Poncelas-Nozal M, Hernando V, Vilardosa Ú, Aluja D, Parra VM, Sanchis D, Garcia-Dorado D. Studies on the role of apoptosis after transient myocardial ischemia: genetic deletion of the executioner caspases-3 and -7 does not limit infarct size and ventricular remodeling. Basic Res Cardiol 111: 18, 2016. doi: 10.1007/s00395-016-0537-6. [DOI] [PubMed] [Google Scholar]
- 207.Irmler M, Thome M, Hahne M, Schneider P, Hofmann K, Steiner V, Bodmer JL, Schröter M, Burns K, Mattmann C, Rimoldi D, French LE, Tschopp J. Inhibition of death receptor signals by cellular FLIP. Nature 388: 190–195, 1997. doi: 10.1038/40657. [DOI] [PubMed] [Google Scholar]
- 208.Iyer SS, He Q, Janczy JR, Elliott EI, Zhong Z, Olivier AK, Sadler JJ, Knepper-Adrian V, Han R, Qiao L, Eisenbarth SC, Nauseef WM, Cassel SL, Sutterwala FS. Mitochondrial cardiolipin is required for Nlrp3 inflammasome activation. Immunity 39: 311–323, 2013. doi: 10.1016/j.immuni.2013.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Jaco I, Annibaldi A, Lalaoui N, Wilson R, Tenev T, Laurien L, Kim C, Jamal K, Wicky John S, Liccardi G, Chau D, Murphy JM, Brumatti G, Feltham R, Pasparakis M, Silke J, Meier P. MK2 Phosphorylates RIPK1 to Prevent TNF-Induced Cell Death. Mol Cell 66: 698–710.e5, 2017. doi: 10.1016/j.molcel.2017.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Jacquemin G, Margiotta D, Kasahara A, Bassoy EY, Walch M, Thiery J, Lieberman J, Martinvalet D. Granzyme B-induced mitochondrial ROS are required for apoptosis. Cell Death Differ 22: 862–874, 2015. doi: 10.1038/cdd.2014.180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Jennings RB, Sommers HM, Herdson PB, Kaltenbach JP. Ischemic injury of myocardium. Ann N Y Acad Sci 156, 1 Experimental: 61–78, 1969. doi: 10.1111/j.1749-6632.1969.tb16718.x. [DOI] [PubMed] [Google Scholar]
- 212.Jennings RB, Sommers HM, Smyth GA, Flack HA, Linn H. Myocardial necrosis induced by temporary occlusion of a coronary artery in the dog. Arch Pathol 70: 68–78, 1960. [PubMed] [Google Scholar]
- 213.Jeremias I, Kupatt C, Martin-Villalba A, Habazettl H, Schenkel J, Boekstegers P, Debatin KM. Involvement of CD95/Apo1/Fas in cell death after myocardial ischemia. Circulation 102: 915–920, 2000. doi: 10.1161/01.CIR.102.8.915. [DOI] [PubMed] [Google Scholar]
- 214.Johnson DB, Balko JM, Compton ML, Chalkias S, Gorham J, Xu Y, Hicks M, Puzanov I, Alexander MR, Bloomer TL, Becker JR, Slosky DA, Phillips EJ, Pilkinton MA, Craig-Owens L, Kola N, Plautz G, Reshef DS, Deutsch JS, Deering RP, Olenchock BA, Lichtman AH, Roden DM, Seidman CE, Koralnik IJ, Seidman JG, Hoffman RD, Taube JM, Diaz LA Jr, Anders RA, Sosman JA, Moslehi JJ. Fulminant Myocarditis with Combination Immune Checkpoint Blockade. N Engl J Med 375: 1749–1755, 2016. doi: 10.1056/NEJMoa1609214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Johnson DB, Chandra S, Sosman JA. Immune Checkpoint Inhibitor Toxicity in 2018. JAMA 320: 1702–1703, 2018. doi: 10.1001/jama.2018.13995. [DOI] [PubMed] [Google Scholar]
- 216.Joiner ML, Koval OM, Li J, He BJ, Allamargot C, Gao Z, Luczak ED, Hall DD, Fink BD, Chen B, Yang J, Moore SA, Scholz TD, Strack S, Mohler PJ, Sivitz WI, Song LS, Anderson ME. CaMKII determines mitochondrial stress responses in heart. Nature 491: 269–273, 2012. doi: 10.1038/nature11444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Juliana C, Fernandes-Alnemri T, Kang S, Farias A, Qin F, Alnemri ES. Non-transcriptional priming and deubiquitination regulate NLRP3 inflammasome activation. J Biol Chem 287: 36617–36622, 2012. doi: 10.1074/jbc.M112.407130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218.Julien O, Wells JA. Caspases and their substrates. Cell Death Differ 24: 1380–1389, 2017. doi: 10.1038/cdd.2017.44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Kaiser WJ, Daley-Bauer LP, Thapa RJ, Mandal P, Berger SB, Huang C, Sundararajan A, Guo H, Roback L, Speck SH, Bertin J, Gough PJ, Balachandran S, Mocarski ES. RIP1 suppresses innate immune necrotic as well as apoptotic cell death during mammalian parturition. Proc Natl Acad Sci USA 111: 7753–7758, 2014. doi: 10.1073/pnas.1401857111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Kaiser WJ, Sridharan H, Huang C, Mandal P, Upton JW, Gough PJ, Sehon CA, Marquis RW, Bertin J, Mocarski ES. Toll-like receptor 3-mediated necrosis via TRIF, RIP3, and MLKL. J Biol Chem 288: 31268–31279, 2013. doi: 10.1074/jbc.M113.462341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221.Kaiser WJ, Upton JW, Long AB, Livingston-Rosanoff D, Daley-Bauer LP, Hakem R, Caspary T, Mocarski ES. RIP3 mediates the embryonic lethality of caspase-8-deficient mice. Nature 471: 368–372, 2011. doi: 10.1038/nature09857. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.Kaiserman D, Bird CH, Sun J, Matthews A, Ung K, Whisstock JC, Thompson PE, Trapani JA, Bird PI. The major human and mouse granzymes are structurally and functionally divergent. J Cell Biol 175: 619–630, 2006. doi: 10.1083/jcb.200606073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Kanayama A, Seth RB, Sun L, Ea CK, Hong M, Shaito A, Chiu YH, Deng L, Chen ZJ. TAB2 and TAB3 activate the NF-kappaB pathway through binding to polyubiquitin chains. Mol Cell 15: 535–548, 2004. doi: 10.1016/j.molcel.2004.08.008. [DOI] [PubMed] [Google Scholar]
- 224.Karbowski M, Norris KL, Cleland MM, Jeong SY, Youle RJ. Role of Bax and Bak in mitochondrial morphogenesis. Nature 443: 658–662, 2006. doi: 10.1038/nature05111. [DOI] [PubMed] [Google Scholar]
- 225.Karch J, Kwong JQ, Burr AR, Sargent MA, Elrod JW, Peixoto PM, Martinez-Caballero S, Osinska H, Cheng EH, Robbins J, Kinnally KW, Molkentin JD. Bax and Bak function as the outer membrane component of the mitochondrial permeability pore in regulating necrotic cell death in mice. eLife 2: e00772, 2013. doi: 10.7554/eLife.00772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226.Karch J, Molkentin JD. Identifying the components of the elusive mitochondrial permeability transition pore. Proc Natl Acad Sci USA 111: 10396–10397, 2014. doi: 10.1073/pnas.1410104111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.Karch J, Schips TG, Maliken BD, Brody MJ, Sargent MA, Kanisicak O, Molkentin JD. Autophagic cell death is dependent on lysosomal membrane permeability through Bax and Bak. eLife 6: e30543, 2017. doi: 10.7554/eLife.30543. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228.Karmazyn M. Amiloride enhances postischemic ventricular recovery: possible role of Na+-H+ exchange. Am J Physiol Heart Circ Physiol 255: H608–H615, 1988. doi: 10.1152/ajpheart.1988.255.3.H608. [DOI] [PubMed] [Google Scholar]
- 229.Kawaguchi M, Takahashi M, Hata T, Kashima Y, Usui F, Morimoto H, Izawa A, Takahashi Y, Masumoto J, Koyama J, Hongo M, Noda T, Nakayama J, Sagara J, Taniguchi S, Ikeda U. Inflammasome activation of cardiac fibroblasts is essential for myocardial ischemia/reperfusion injury. Circulation 123: 594–604, 2011. doi: 10.1161/CIRCULATIONAHA.110.982777. [DOI] [PubMed] [Google Scholar]
- 230.Kayagaki N, Stowe IB, Lee BL, O’Rourke K, Anderson K, Warming S, Cuellar T, Haley B, Roose-Girma M, Phung QT, Liu PS, Lill JR, Li H, Wu J, Kummerfeld S, Zhang J, Lee WP, Snipas SJ, Salvesen GS, Morris LX, Fitzgerald L, Zhang Y, Bertram EM, Goodnow CC, Dixit VM. Caspase-11 cleaves gasdermin D for non-canonical inflammasome signalling. Nature 526: 666–671, 2015. doi: 10.1038/nature15541. [DOI] [PubMed] [Google Scholar]
- 231.Kayagaki N, Warming S, Lamkanfi M, Vande Walle L, Louie S, Dong J, Newton K, Qu Y, Liu J, Heldens S, Zhang J, Lee WP, Roose-Girma M, Dixit VM. Non-canonical inflammasome activation targets caspase-11. Nature 479: 117–121, 2011. doi: 10.1038/nature10558. [DOI] [PubMed] [Google Scholar]
- 232.Ke FFS, Vanyai HK, Cowan AD, Delbridge ARD, Whitehead L, Grabow S, Czabotar PE, Voss AK, Strasser A. Embryogenesis and Adult Life in the Absence of Intrinsic Apoptosis Effectors BAX, BAK, and BOK. Cell 173: 1217–1230.e17, 2018. doi: 10.1016/j.cell.2018.04.036. [DOI] [PubMed] [Google Scholar]
- 233.Kelliher MA, Grimm S, Ishida Y, Kuo F, Stanger BZ, Leder P. The death domain kinase RIP mediates the TNF-induced NF-kappaB signal. Immunity 8: 297–303, 1998. doi: 10.1016/S1074-7613(00)80535-X. [DOI] [PubMed] [Google Scholar]
- 234.Kerr JF. Shrinkage necrosis: a distinct mode of cellular death. J Pathol 105: 13–20, 1971. doi: 10.1002/path.1711050103. [DOI] [PubMed] [Google Scholar]
- 235.Kerr JF, Wyllie AH, Currie AR. Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics. Br J Cancer 26: 239–257, 1972. doi: 10.1038/bjc.1972.33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236.Kersse K, Verspurten J, Vanden Berghe T, Vandenabeele P. The death-fold superfamily of homotypic interaction motifs. Trends Biochem Sci 36: 541–552, 2011. doi: 10.1016/j.tibs.2011.06.006. [DOI] [PubMed] [Google Scholar]
- 237.Khattri R, Auger JA, Griffin MD, Sharpe AH, Bluestone JA. Lymphoproliferative disorder in CTLA-4 knockout mice is characterized by CD28-regulated activation of Th2 responses. J Immunol 162: 5784–5791, 1999. [PubMed] [Google Scholar]
- 238.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240.Konstantinidis K, Whelan RS, Kitsis RN. Mechanisms of cell death in heart disease. Arterioscler Thromb Vasc Biol 32: 1552–1562, 2012. doi: 10.1161/ATVBAHA.111.224915. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 241.Koseki T, Inohara N, Chen S, Núñez G. ARC, an inhibitor of apoptosis expressed in skeletal muscle and heart that interacts selectively with caspases. Proc Natl Acad Sci USA 95: 5156–5160, 1998. doi: 10.1073/pnas.95.9.5156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242.Koudstaal S, Oerlemans MI, Van der Spoel TI, Janssen AW, Hoefer IE, Doevendans PA, Sluijter JP, Chamuleau SA. Necrostatin-1 alleviates reperfusion injury following acute myocardial infarction in pigs. Eur J Clin Invest 45: 150–159, 2015. doi: 10.1111/eci.12391. [DOI] [PubMed] [Google Scholar]
- 243.Kovacsovics M, Martinon F, Micheau O, Bodmer JL, Hofmann K, Tschopp J. Overexpression of Helicard, a CARD-containing helicase cleaved during apoptosis, accelerates DNA degradation. Curr Biol 12: 838–843, 2002. doi: 10.1016/S0960-9822(02)00842-4. [DOI] [PubMed] [Google Scholar]
- 244.Kovalenko A, Chable-Bessia C, Cantarella G, Israël A, Wallach D, Courtois G. The tumour suppressor CYLD negatively regulates NF-kappaB signalling by deubiquitination. Nature 424: 801–805, 2003. doi: 10.1038/nature01802. [DOI] [PubMed] [Google Scholar]
- 245.Kraus WL. Transcriptional control by PARP-1: chromatin modulation, enhancer-binding, coregulation, and insulation. Curr Opin Cell Biol 20: 294–302, 2008. doi: 10.1016/j.ceb.2008.03.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 246.Kreuz S, Siegmund D, Scheurich P, Wajant H. NF-kappaB inducers upregulate cFLIP, a cycloheximide-sensitive inhibitor of death receptor signaling. Mol Cell Biol 21: 3964–3973, 2001. doi: 10.1128/MCB.21.12.3964-3973.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247.Krishna S, Overholtzer M. Mechanisms and consequences of entosis. Cell Mol Life Sci 73: 2379–2386, 2016. doi: 10.1007/s00018-016-2207-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248.Kung G, Konstantinidis K, Kitsis RN. Programmed necrosis, not apoptosis, in the heart. Circ Res 108: 1017–1036, 2011. doi: 10.1161/CIRCRESAHA.110.225730. [DOI] [PubMed] [Google Scholar]
- 249.Kupka S, De Miguel D, Draber P, Martino L, Surinova S, Rittinger K, Walczak H. SPATA2-Mediated Binding of CYLD to HOIP Enables CYLD Recruitment to Signaling Complexes. Cell Rep 16: 2271–2280, 2016. doi: 10.1016/j.celrep.2016.07.086. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250.Kurrelmeyer KM, Michael LH, Baumgarten G, Taffet GE, Peschon JJ, Sivasubramanian N, Entman ML, Mann DL. Endogenous tumor necrosis factor protects the adult cardiac myocyte against ischemic-induced apoptosis in a murine model of acute myocardial infarction. Proc Natl Acad Sci USA 97: 5456–5461, 2000. doi: 10.1073/pnas.070036297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251.Kwong JQ, Davis J, Baines CP, Sargent MA, Karch J, Wang X, Huang T, Molkentin JD. Genetic deletion of the mitochondrial phosphate carrier desensitizes the mitochondrial permeability transition pore and causes cardiomyopathy. Cell Death Differ 21: 1209–1217, 2014. doi: 10.1038/cdd.2014.36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 252.Lafont E, Draber P, Rieser E, Reichert M, Kupka S, de Miguel D, Draberova H, von Mässenhausen A, Bhamra A, Henderson S, Wojdyla K, Chalk A, Surinova S, Linkermann A, Walczak H. TBK1 and IKKε prevent TNF-induced cell death by RIPK1 phosphorylation. Nat Cell Biol 20: 1389–1399, 2018. doi: 10.1038/s41556-018-0229-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 253.Lanier LL, O’Fallon S, Somoza C, Phillips JH, Linsley PS, Okumura K, Ito D, Azuma M. CD80 (B7) and CD86 (B70) provide similar costimulatory signals for T cell proliferation, cytokine production, and generation of CTL. J Immunol 154: 97–105, 1995. [PubMed] [Google Scholar]
- 254.Latchman Y, Wood CR, Chernova T, Chaudhary D, Borde M, Chernova I, Iwai Y, Long AJ, Brown JA, Nunes R, Greenfield EA, Bourque K, Boussiotis VA, Carter LL, Carreno BM, Malenkovich N, Nishimura H, Okazaki T, Honjo T, Sharpe AH, Freeman GJ. PD-L2 is a second ligand for PD-1 and inhibits T cell activation. Nat Immunol 2: 261–268, 2001. doi: 10.1038/85330. [DOI] [PubMed] [Google Scholar]
- 255.Latchman YE, Liang SC, Wu Y, Chernova T, Sobel RA, Klemm M, Kuchroo VK, Freeman GJ, Sharpe AH. PD-L1-deficient mice show that PD-L1 on T cells, antigen-presenting cells, and host tissues negatively regulates T cells. Proc Natl Acad Sci USA 101: 10691–10696, 2004. doi: 10.1073/pnas.0307252101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 256.Latz E, Xiao TS, Stutz A. Activation and regulation of the inflammasomes. Nat Rev Immunol 13: 397–411, 2013. doi: 10.1038/nri3452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 257.Lavrik IN, Krammer PH. Regulation of CD95/Fas signaling at the DISC. Cell Death Differ 19: 36–41, 2012. doi: 10.1038/cdd.2011.155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 258.Lee P, Sata M, Lefer DJ, Factor SM, Walsh K, Kitsis RN. Fas pathway is a critical mediator of cardiac myocyte death and MI during ischemia-reperfusion in vivo. Am J Physiol Heart Circ Physiol 284: H456–H463, 2003. doi: 10.1152/ajpheart.00777.2002. [DOI] [PubMed] [Google Scholar]
- 259.Lee TH, Shank J, Cusson N, Kelliher MA. The kinase activity of Rip1 is not required for tumor necrosis factor-alpha-induced IkappaB kinase or p38 MAP kinase activation or for the ubiquitination of Rip1 by Traf2. J Biol Chem 279: 33185–33191, 2004. doi: 10.1074/jbc.M404206200. [DOI] [PubMed] [Google Scholar]
- 260.Leist M, Single B, Castoldi AF, Kühnle S, Nicotera P. Intracellular adenosine triphosphate (ATP) concentration: a switch in the decision between apoptosis and necrosis. J Exp Med 185: 1481–1486, 1997. doi: 10.1084/jem.185.8.1481. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 261.Levine B, Yuan J. Autophagy in cell death: an innocent convict? J Clin Invest 115: 2679–2688, 2005. doi: 10.1172/JCI26390. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 262.Li D, Xu T, Cao Y, Wang H, Li L, Chen S, Wang X, Shen Z. A cytosolic heat shock protein 90 and cochaperone CDC37 complex is required for RIP3 activation during necroptosis. Proc Natl Acad Sci USA 112: 5017–5022, 2015. doi: 10.1073/pnas.1505244112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 263.Li H, Zhu H, Xu CJ, Yuan J. Cleavage of BID by caspase 8 mediates the mitochondrial damage in the Fas pathway of apoptosis. Cell 94: 491–501, 1998. doi: 10.1016/S0092-8674(00)81590-1. [DOI] [PubMed] [Google Scholar]
- 264.Li J, McQuade T, Siemer AB, Napetschnig J, Moriwaki K, Hsiao YS, Damko E, Moquin D, Walz T, McDermott A, Chan FK, Wu H. The RIP1/RIP3 necrosome forms a functional amyloid signaling complex required for programmed necrosis. Cell 150: 339–350, 2012. doi: 10.1016/j.cell.2012.06.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 265.Li L, Chen Y, Doan J, Murray J, Molkentin JD, Liu Q. Transforming growth factor β-activated kinase 1 signaling pathway critically regulates myocardial survival and remodeling. Circulation 130: 2162–2172, 2014. doi: 10.1161/CIRCULATIONAHA.114.011195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 266.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. [DOI] [PubMed] [Google Scholar]
- 267.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. [DOI] [PubMed] [Google Scholar]
- 268.Li W, Feng G, Gauthier JM, Lokshina I, Higashikubo R, Evans S, Liu X, Hassan A, Tanaka S, Cicka M, Hsiao HM, Ruiz-Perez D, Bredemeyer A, Gross RW, Mann DL, Tyurina YY, Gelman AE, Kagan VE, Linkermann A, Lavine KJ, Kreisel D. Ferroptotic cell death and TLR4/Trif signaling initiate neutrophil recruitment after heart transplantation. J Clin Invest 129: 2293–2304, 2019. doi: 10.1172/JCI126428. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 269.Liaudet L, Szabó E, Timashpolsky L, Virág L, Cziráki A, Szabó C. Suppression of poly (ADP-ribose) polymerase activation by 3-aminobenzamide in a rat model of myocardial infarction: long-term morphological and functional consequences. Br J Pharmacol 133: 1424–1430, 2001. doi: 10.1038/sj.bjp.0704185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 270.Libby P, Pasterkamp G. Requiem for the ‟vulnerable plaqueˮ. Eur Heart J 36: 2984–2987, 2015. [DOI] [PubMed] [Google Scholar]
- 271.Libby P, Theroux P. Pathophysiology of coronary artery disease. Circulation 111: 3481–3488, 2005. doi: 10.1161/CIRCULATIONAHA.105.537878. [DOI] [PubMed] [Google Scholar]
- 272.Lim SY, Davidson SM, Mocanu MM, Yellon DM, Smith CC. The cardioprotective effect of necrostatin requires the cyclophilin-D component of the mitochondrial permeability transition pore. Cardiovasc Drugs Ther 21: 467–469, 2007. doi: 10.1007/s10557-007-6067-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 273.Lim WY, Messow CM, Berry C. Cyclosporin variably and inconsistently reduces infarct size in experimental models of reperfused myocardial infarction: a systematic review and meta-analysis. Br J Pharmacol 165: 2034–2043, 2012. doi: 10.1111/j.1476-5381.2011.01691.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 274.Lin J, Kumari S, Kim C, Van TM, Wachsmuth L, Polykratis A, Pasparakis M. RIPK1 counteracts ZBP1-mediated necroptosis to inhibit inflammation. Nature 540: 124–128, 2016. doi: 10.1038/nature20558. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 275.Lin KM, Hu W, Troutman TD, Jennings M, Brewer T, Li X, Nanda S, Cohen P, Thomas JA, Pasare C. IRAK-1 bypasses priming and directly links TLRs to rapid NLRP3 inflammasome activation. [Correction in Proc Natl Acad Sci USA 111: 3195, 2014.] Proc Natl Acad Sci USA 111: 775–780, 2014. doi: 10.1073/pnas.1320294111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 276.Lin Y, Devin A, Rodriguez Y, Liu ZG. Cleavage of the death domain kinase RIP by caspase-8 prompts TNF-induced apoptosis. Genes Dev 13: 2514–2526, 1999. doi: 10.1101/gad.13.19.2514. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 277.Linkermann A, Green DR. Necroptosis. N Engl J Med 370: 455–465, 2014. doi: 10.1056/NEJMra1310050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 278.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 279.Linkermann A, Skouta R, Himmerkus N, Mulay SR, Dewitz C, De Zen F, Prokai A, Zuchtriegel G, Krombach F, Welz PS, Weinlich R, Vanden Berghe T, Vandenabeele P, Pasparakis M, Bleich M, Weinberg JM, Reichel CA, Bräsen JH, Kunzendorf U, Anders HJ, Stockwell BR, Green DR, Krautwald S. Synchronized renal tubular cell death involves ferroptosis. Proc Natl Acad Sci USA 111: 16836–16841, 2014. doi: 10.1073/pnas.1415518111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 280.Linsley PS, Brady W, Urnes M, Grosmaire LS, Damle NK, Ledbetter JA. CTLA-4 is a second receptor for the B cell activation antigen B7. J Exp Med 174: 561–569, 1991. doi: 10.1084/jem.174.3.561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 281.Linsley PS, Greene JL, Brady W, Bajorath J, Ledbetter JA, Peach R. Human B7-1 (CD80) and B7-2 (CD86) bind with similar avidities but distinct kinetics to CD28 and CTLA-4 receptors. Immunity 1: 793–801, 1994. doi: 10.1016/S1074-7613(94)80021-9. [DOI] [PubMed] [Google Scholar]
- 282.Liu HR, Gao E, Hu A, Tao L, Qu Y, Most P, Koch WJ, Christopher TA, Lopez BL, Alnemri ES, Zervos AS, Ma XL. Role of Omi/HtrA2 in apoptotic cell death after myocardial ischemia and reperfusion. Circulation 111: 90–96, 2005. doi: 10.1161/01.CIR.0000151613.90994.17. [DOI] [PubMed] [Google Scholar]
- 283.Liu S, Liu H, Johnston A, Hanna-Addams S, Reynoso E, Xiang Y, Wang Z. MLKL forms disulfide bond-dependent amyloid-like polymers to induce necroptosis. Proc Natl Acad Sci USA 114: E7450–E7459, 2017. doi: 10.1073/pnas.1707531114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 284.Liu X, Kim CN, Yang J, Jemmerson R, Wang X. Induction of apoptotic program in cell-free extracts: requirement for dATP and cytochrome c. Cell 86: 147–157, 1996. doi: 10.1016/S0092-8674(00)80085-9. [DOI] [PubMed] [Google Scholar]
- 285.Liu X, Li P, Widlak P, Zou H, Luo X, Garrard WT, Wang X. The 40-kDa subunit of DNA fragmentation factor induces DNA fragmentation and chromatin condensation during apoptosis. Proc Natl Acad Sci USA 95: 8461–8466, 1998. doi: 10.1073/pnas.95.15.8461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 286.Liu X, Zhang Z, Ruan J, Pan Y, Magupalli VG, Wu H, Lieberman J. Inflammasome-activated gasdermin D causes pyroptosis by forming membrane pores. Nature 535: 153–158, 2016. doi: 10.1038/nature18629. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 287.Liu X, Zou H, Slaughter C, Wang X. DFF, a heterodimeric protein that functions downstream of caspase-3 to trigger DNA fragmentation during apoptosis. Cell 89: 175–184, 1997. doi: 10.1016/S0092-8674(00)80197-X. [DOI] [PubMed] [Google Scholar]
- 288.Liu Y, Fan C, Zhang Y, Yu X, Wu X, Zhang X, Zhao Q, Zhang H, Xie Q, Li M, Li X, Ding Q, Ying H, Li D, Zhang H. RIP1 kinase activity-dependent roles in embryonic development of Fadd-deficient mice. Cell Death Differ 24: 1459–1469, 2017. doi: 10.1038/cdd.2017.78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 289.Liu Y, Levine B. Autosis and autophagic cell death: the dark side of autophagy. Cell Death Differ 22: 367–376, 2015. doi: 10.1038/cdd.2014.143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 290.Liu Y, Shoji-Kawata S, Sumpter RM Jr, Wei Y, Ginet V, Zhang L, Posner B, Tran KA, Green DR, Xavier RJ, Shaw SY, Clarke PG, Puyal J, Levine B. Autosis is a Na+,K+-ATPase-regulated form of cell death triggered by autophagy-inducing peptides, starvation, and hypoxia-ischemia. Proc Natl Acad Sci USA 110: 20364–20371, 2013. doi: 10.1073/pnas.1319661110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 291.Liu ZG, Hsu H, Goeddel DV, Karin M. Dissection of TNF receptor 1 effector functions: JNK activation is not linked to apoptosis while NF-kappaB activation prevents cell death. Cell 87: 565–576, 1996. doi: 10.1016/S0092-8674(00)81375-6. [DOI] [PubMed] [Google Scholar]
- 292.Llambi F, Moldoveanu T, Tait SW, Bouchier-Hayes L, Temirov J, McCormick LL, Dillon CP, Green DR. A unified model of mammalian BCL-2 protein family interactions at the mitochondria. Mol Cell 44: 517–531, 2011. doi: 10.1016/j.molcel.2011.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 293.Llambi F, Wang YM, Victor B, Yang M, Schneider DM, Gingras S, Parsons MJ, Zheng JH, Brown SA, Pelletier S, Moldoveanu T, Chen T, Green DR. BOK Is a Non-canonical BCL-2 Family Effector of Apoptosis Regulated by ER-Associated Degradation. Cell 165: 421–433, 2016. doi: 10.1016/j.cell.2016.02.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 294.Lockshin RA, Williams CM. Programmed Cell Death. I. Cytology of Degeneration in the Intersegmental Muscles of the Pernyi Silkmoth. J Insect Physiol 11: 123–133, 1965. doi: 10.1016/0022-1910(65)90099-5. [DOI] [PubMed] [Google Scholar]
- 295.Lörincz T, Jemnitz K, Kardon T, Mandl J, Szarka A. Ferroptosis is Involved in Acetaminophen Induced Cell Death. Pathol Oncol Res 21: 1115–1121, 2015. doi: 10.1007/s12253-015-9946-3. [DOI] [PubMed] [Google Scholar]
- 296.Lu A, Magupalli VG, Ruan J, Yin Q, Atianand MK, Vos MR, Schröder GF, Fitzgerald KA, Wu H, Egelman EH. Unified polymerization mechanism for the assembly of ASC-dependent inflammasomes. Cell 156: 1193–1206, 2014. doi: 10.1016/j.cell.2014.02.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 297.Lucchesi BR. Modulation of leukocyte-mediated myocardial reperfusion injury. Annu Rev Physiol 52: 561–576, 1990. doi: 10.1146/annurev.ph.52.030190.003021. [DOI] [PubMed] [Google Scholar]
- 298.Luedde M, Lutz M, Carter N, Sosna J, Jacoby C, Vucur M, Gautheron J, Roderburg C, Borg N, Reisinger F, Hippe HJ, Linkermann A, Wolf MJ, Rose-John S, Lüllmann-Rauch R, Adam D, Flögel U, Heikenwalder M, Luedde T, Frey N. RIP3, a kinase promoting necroptotic cell death, mediates adverse remodelling after myocardial infarction. Cardiovasc Res 103: 206–216, 2014. doi: 10.1093/cvr/cvu146. [DOI] [PubMed] [Google Scholar]
- 299.Luo X, Budihardjo I, Zou H, Slaughter C, Wang X. Bid, a Bcl2 interacting protein, mediates cytochrome c release from mitochondria in response to activation of cell surface death receptors. Cell 94: 481–490, 1998. doi: 10.1016/S0092-8674(00)81589-5. [DOI] [PubMed] [Google Scholar]
- 300.Maejima Y, Kyoi S, Zhai P, Liu T, Li H, Ivessa A, Sciarretta S, Del Re DP, Zablocki DK, Hsu CP, Lim DS, Isobe M, Sadoshima J. Mst1 inhibits autophagy by promoting the interaction between Beclin1 and Bcl-2. Nat Med 19: 1478–1488, 2013. doi: 10.1038/nm.3322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 301.Mahmood SS, Fradley MG, Cohen JV, Nohria A, Reynolds KL, Heinzerling LM, Sullivan RJ, Damrongwatanasuk R, Chen CL, Gupta D, Kirchberger MC, Awadalla M, Hassan MZO, Moslehi JJ, Shah SP, Ganatra S, Thavendiranathan P, Lawrence DP, Groarke JD, Neilan TG. Myocarditis in Patients Treated With Immune Checkpoint Inhibitors. J Am Coll Cardiol 71: 1755–1764, 2018. doi: 10.1016/j.jacc.2018.02.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 302.Malin JZ, Shaham S. Cell Death in C. elegans Development. Curr Top Dev Biol 114: 1–42, 2015. doi: 10.1016/bs.ctdb.2015.07.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 303.Mandal P, Berger SB, Pillay S, Moriwaki K, Huang C, Guo H, Lich JD, Finger J, Kasparcova V, Votta B, Ouellette M, King BW, Wisnoski D, Lakdawala AS, DeMartino MP, Casillas LN, Haile PA, Sehon CA, Marquis RW, Upton J, Daley-Bauer LP, Roback L, Ramia N, Dovey CM, Carette JE, Chan FK, Bertin J, Gough PJ, Mocarski ES, Kaiser WJ. RIP3 induces apoptosis independent of pronecrotic kinase activity. Mol Cell 56: 481–495, 2014. doi: 10.1016/j.molcel.2014.10.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 304.Martínez-Lostao L, Anel A, Pardo J. How Do Cytotoxic Lymphocytes Kill Cancer Cells? Clin Cancer Res 21: 5047–5056, 2015. doi: 10.1158/1078-0432.CCR-15-0685. [DOI] [PubMed] [Google Scholar]
- 305.Martinon F, Burns K, Tschopp J. The inflammasome: a molecular platform triggering activation of inflammatory caspases and processing of proIL-beta. Mol Cell 10: 417–426, 2002. doi: 10.1016/S1097-2765(02)00599-3. [DOI] [PubMed] [Google Scholar]
- 306.Martinou JC, Youle RJ. Mitochondria in apoptosis: Bcl-2 family members and mitochondrial dynamics. Dev Cell 21: 92–101, 2011. doi: 10.1016/j.devcel.2011.06.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 307.Mashimo M, Kato J, Moss J. ADP-ribosyl-acceptor hydrolase 3 regulates poly (ADP-ribose) degradation and cell death during oxidative stress. Proc Natl Acad Sci USA 110: 18964–18969, 2013. doi: 10.1073/pnas.1312783110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 308.Matsuda T, Zhai P, Sciarretta S, Zhang Y, Jeong JI, Ikeda S, Park J, Hsu CP, Tian B, Pan D, Sadoshima J, Del Re DP. NF2 Activates Hippo Signaling and Promotes Ischemia/Reperfusion Injury in the Heart. Circ Res 119: 596–606, 2016. doi: 10.1161/CIRCRESAHA.116.308586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 309.Matsui T, Tao J, del Monte F, Lee KH, Li L, Picard M, Force TL, Franke TF, Hajjar RJ, Rosenzweig A. Akt activation preserves cardiac function and prevents injury after transient cardiac ischemia in vivo. Circulation 104: 330–335, 2001. doi: 10.1161/01.CIR.104.3.330. [DOI] [PubMed] [Google Scholar]
- 310.Matsui Y, Takagi H, Qu X, Abdellatif M, Sakoda H, Asano T, Levine B, Sadoshima J. Distinct roles of autophagy in the heart during ischemia and reperfusion: roles of AMP-activated protein kinase and Beclin 1 in mediating autophagy. Circ Res 100: 914–922, 2007. doi: 10.1161/01.RES.0000261924.76669.36. [DOI] [PubMed] [Google Scholar]
- 311.Matsushima M, Fujiwara T, Takahashi E, Minaguchi T, Eguchi Y, Tsujimoto Y, Suzumori K, Nakamura Y. Isolation, mapping, and functional analysis of a novel human cDNA (BNIP3L) encoding a protein homologous to human NIP3. Genes Chromosomes Cancer 21: 230–235, 1998. doi:. [DOI] [PubMed] [Google Scholar]
- 312.McArthur K, Kile BT. Apoptotic Caspases: Multiple or Mistaken Identities? Trends Cell Biol 28: 475–493, 2018. doi: 10.1016/j.tcb.2018.02.003. [DOI] [PubMed] [Google Scholar]
- 313.McKimpson WM, Weinberger J, Czerski L, Zheng M, Crow MT, Pessin JE, Chua SC Jr, Kitsis RN. The apoptosis inhibitor ARC alleviates the ER stress response to promote β-cell survival. Diabetes 62: 183–193, 2013. doi: 10.2337/db12-0504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 314.Mehlen P. Dependence receptors: the trophic theory revisited. Sci Signal 3: pe47, 2010. doi: 10.1126/scisignal.3151pe47. [DOI] [PubMed] [Google Scholar]
- 315.Meng H, Liu Z, Li X, Wang H, Jin T, Wu G, Shan B, Christofferson DE, Qi C, Yu Q, Li Y, Yuan J. Death-domain dimerization-mediated activation of RIPK1 controls necroptosis and RIPK1-dependent apoptosis. Proc Natl Acad Sci USA 115: E2001–E2009, 2018. doi: 10.1073/pnas.1722013115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 316.Menon MB, Gropengießer J, Fischer J, Novikova L, Deuretzbacher A, Lafera J, Schimmeck H, Czymmeck N, Ronkina N, Kotlyarov A, Aepfelbacher M, Gaestel M, Ruckdeschel K. p38MAPK/MK2-dependent phosphorylation controls cytotoxic RIPK1 signalling in inflammation and infection. Nat Cell Biol 19: 1248–1259, 2017. doi: 10.1038/ncb3614. [DOI] [PubMed] [Google Scholar]
- 317.Merkle S, Frantz S, Schön MP, Bauersachs J, Buitrago M, Frost RJ, Schmitteckert EM, Lohse MJ, Engelhardt S. A role for caspase-1 in heart failure. Circ Res 100: 645–653, 2007. doi: 10.1161/01.RES.0000260203.55077.61. [DOI] [PubMed] [Google Scholar]
- 318.Metkar SS, Wang B, Ebbs ML, Kim JH, Lee YJ, Raja SM, Froelich CJ. Granzyme B activates procaspase-3 which signals a mitochondrial amplification loop for maximal apoptosis. J Cell Biol 160: 875–885, 2003. doi: 10.1083/jcb.200210158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 319.Mezzaroma E, Toldo S, Farkas D, Seropian IM, Van Tassell BW, Salloum FN, Kannan HR, Menna AC, Voelkel NF, Abbate A. The inflammasome promotes adverse cardiac remodeling following acute myocardial infarction in the mouse. Proc Natl Acad Sci USA 108: 19725–19730, 2011. doi: 10.1073/pnas.1108586108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 320.Miao W, Luo Z, Kitsis RN, Walsh K. Intracoronary, adenovirus-mediated Akt gene transfer in heart limits infarct size following ischemia-reperfusion injury in vivo. J Mol Cell Cardiol 32: 2397–2402, 2000. doi: 10.1006/jmcc.2000.1283. [DOI] [PubMed] [Google Scholar]
- 321.Michaud M, Martins I, Sukkurwala AQ, Adjemian S, Ma Y, Pellegatti P, Shen S, Kepp O, Scoazec M, Mignot G, Rello-Varona S, Tailler M, Menger L, Vacchelli E, Galluzzi L, Ghiringhelli F, di Virgilio F, Zitvogel L, Kroemer G. Autophagy-dependent anticancer immune responses induced by chemotherapeutic agents in mice. Science 334: 1573–1577, 2011. doi: 10.1126/science.1208347. [DOI] [PubMed] [Google Scholar]
- 322.Micheau O, Lens S, Gaide O, Alevizopoulos K, Tschopp J. NF-kappaB signals induce the expression of c-FLIP. Mol Cell Biol 21: 5299–5305, 2001. doi: 10.1128/MCB.21.16.5299-5305.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 323.Micheau O, Tschopp J. Induction of TNF receptor I-mediated apoptosis via two sequential signaling complexes. Cell 114: 181–190, 2003. doi: 10.1016/S0092-8674(03)00521-X. [DOI] [PubMed] [Google Scholar]
- 324.Miller EJ, Li J, Leng L, McDonald C, Atsumi T, Bucala R, Young LH. Macrophage migration inhibitory factor stimulates AMP-activated protein kinase in the ischaemic heart. Nature 451: 578–582, 2008. doi: 10.1038/nature06504. [DOI] [PubMed] [Google Scholar]
- 325.Miyashita T, Reed JC. Tumor suppressor p53 is a direct transcriptional activator of the human bax gene. Cell 80: 293–299, 1995. doi: 10.1016/0092-8674(95)90412-3. [DOI] [PubMed] [Google Scholar]
- 326.Mizushima N, Komatsu M. Autophagy: renovation of cells and tissues. Cell 147: 728–741, 2011. doi: 10.1016/j.cell.2011.10.026. [DOI] [PubMed] [Google Scholar]
- 327.Moe GW, Marín-García J. Role of cell death in the progression of heart failure. Heart Fail Rev 21: 157–167, 2016. doi: 10.1007/s10741-016-9532-0. [DOI] [PubMed] [Google Scholar]
- 328.Monden Y, Kubota T, Inoue T, Tsutsumi T, Kawano S, Ide T, Tsutsui H, Sunagawa K. Tumor necrosis factor-alpha is toxic via receptor 1 and protective via receptor 2 in a murine model of myocardial infarction. Am J Physiol Heart Circ Physiol 293: H743–H753, 2007. doi: 10.1152/ajpheart.00166.2007. [DOI] [PubMed] [Google Scholar]
- 329.Montessuit S, Somasekharan SP, Terrones O, Lucken-Ardjomande S, Herzig S, Schwarzenbacher R, Manstein DJ, Bossy-Wetzel E, Basañez G, Meda P, Martinou JC. Membrane remodeling induced by the dynamin-related protein Drp1 stimulates Bax oligomerization. Cell 142: 889–901, 2010. doi: 10.1016/j.cell.2010.08.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 330.Morrow DA, Brickman CM, Murphy SA, Baran K, Krakover R, Dauerman H, Kumar S, Slomowitz N, Grip L, McCabe CH, Salzman AL. A randomized, placebo-controlled trial to evaluate the tolerability, safety, pharmacokinetics, and pharmacodynamics of a potent inhibitor of poly(ADP-ribose) polymerase (INO-1001) in patients with ST-elevation myocardial infarction undergoing primary percutaneous coronary intervention: results of the TIMI 37 trial. J Thromb Thrombolysis 27: 359–364, 2009. doi: 10.1007/s11239-008-0230-1. [DOI] [PubMed] [Google Scholar]
- 331.Moslehi J, Amgalan D, Kitsis RN. Grounding Cardio-Oncology in Basic and Clinical Science. Circulation 136: 3–5, 2017. doi: 10.1161/CIRCULATIONAHA.117.025393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 332.Moslehi JJ. Cardiovascular Toxic Effects of Targeted Cancer Therapies. N Engl J Med 375: 1457–1467, 2016. doi: 10.1056/NEJMra1100265. [DOI] [PubMed] [Google Scholar]
- 333.Moslehi JJ, Salem JE, Sosman JA, Lebrun-Vignes B, Johnson DB. Increased reporting of fatal immune checkpoint inhibitor-associated myocarditis. Lancet 391: 933, 2018. doi: 10.1016/S0140-6736(18)30533-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 334.Moubarak RS, Yuste VJ, Artus C, Bouharrour A, Greer PA, Menissier-de Murcia J, Susin SA. Sequential activation of poly(ADP-ribose) polymerase 1, calpains, and Bax is essential in apoptosis-inducing factor-mediated programmed necrosis. Mol Cell Biol 27: 4844–4862, 2007. doi: 10.1128/MCB.02141-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 335.Murphy E, Perlman M, London RE, Steenbergen C. Amiloride delays the ischemia-induced rise in cytosolic free calcium. Circ Res 68: 1250–1258, 1991. doi: 10.1161/01.RES.68.5.1250. [DOI] [PubMed] [Google Scholar]
- 336.Murphy E, Steenbergen C. Mechanisms underlying acute protection from cardiac ischemia-reperfusion injury. Physiol Rev 88: 581–609, 2008. doi: 10.1152/physrev.00024.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 337.Murphy JM, Czabotar PE, Hildebrand JM, Lucet IS, Zhang JG, Alvarez-Diaz S, Lewis R, Lalaoui N, Metcalf D, Webb AI, Young SN, Varghese LN, Tannahill GM, Hatchell EC, Majewski IJ, Okamoto T, Dobson RC, Hilton DJ, Babon JJ, Nicola NA, Strasser A, Silke J, Alexander WS. The pseudokinase MLKL mediates necroptosis via a molecular switch mechanism. Immunity 39: 443–453, 2013. doi: 10.1016/j.immuni.2013.06.018. [DOI] [PubMed] [Google Scholar]
- 338.Muzio M, Chinnaiyan AM, Kischkel FC, O’Rourke K, Shevchenko A, Ni J, Scaffidi C, Bretz JD, Zhang M, Gentz R, Mann M, Krammer PH, Peter ME, Dixit VM. FLICE, a novel FADD-homologous ICE/CED-3-like protease, is recruited to the CD95 (Fas/APO-1) death-inducing signaling complex. Cell 85: 817–827, 1996. doi: 10.1016/S0092-8674(00)81266-0. [DOI] [PubMed] [Google Scholar]
- 339.Muzio M, Stockwell BR, Stennicke HR, Salvesen GS, Dixit VM. An induced proximity model for caspase-8 activation. J Biol Chem 273: 2926–2930, 1998. doi: 10.1074/jbc.273.5.2926. [DOI] [PubMed] [Google Scholar]
- 340.Nabel EG, Braunwald E. A tale of coronary artery disease and myocardial infarction. N Engl J Med 366: 54–63, 2012. doi: 10.1056/NEJMra1112570. [DOI] [PubMed] [Google Scholar]
- 341.Nakagawa T, Shimizu S, Watanabe T, Yamaguchi O, Otsu K, Yamagata H, Inohara H, Kubo T, Tsujimoto Y. Cyclophilin D-dependent mitochondrial permeability transition regulates some necrotic but not apoptotic cell death. Nature 434: 652–658, 2005. doi: 10.1038/nature03317. [DOI] [PubMed] [Google Scholar]
- 343.Nakahira K, Haspel JA, Rathinam VA, Lee SJ, Dolinay T, Lam HC, Englert JA, Rabinovitch M, Cernadas M, Kim HP, Fitzgerald KA, Ryter SW, Choi AM. Autophagy proteins regulate innate immune responses by inhibiting the release of mitochondrial DNA mediated by the NALP3 inflammasome. Nat Immunol 12: 222–230, 2011. doi: 10.1038/ni.1980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 344.Nakai A, Yamaguchi O, Takeda T, Higuchi Y, Hikoso S, Taniike M, Omiya S, Mizote I, Matsumura Y, Asahi M, Nishida K, Hori M, Mizushima N, Otsu K. The role of autophagy in cardiomyocytes in the basal state and in response to hemodynamic stress. Nat Med 13: 619–624, 2007. doi: 10.1038/nm1574. [DOI] [PubMed] [Google Scholar]
- 345.Nakano K, Vousden KH. PUMA, a novel proapoptotic gene, is induced by p53. Mol Cell 7: 683–694, 2001. doi: 10.1016/S1097-2765(01)00214-3. [DOI] [PubMed] [Google Scholar]
- 346.Nakayama H, Chen X, Baines CP, Klevitsky R, Zhang X, Zhang H, Jaleel N, Chua BH, Hewett TE, Robbins J, Houser SR, Molkentin JD. Ca2+- and mitochondrial-dependent cardiomyocyte necrosis as a primary mediator of heart failure. J Clin Invest 117: 2431–2444, 2007. doi: 10.1172/JCI31060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 347.Nam YJ, Mani K, Ashton AW, Peng CF, Krishnamurthy B, Hayakawa Y, Lee P, Korsmeyer SJ, Kitsis RN. Inhibition of both the extrinsic and intrinsic death pathways through nonhomotypic death-fold interactions. Mol Cell 15: 901–912, 2004. doi: 10.1016/j.molcel.2004.08.020. [DOI] [PubMed] [Google Scholar]
- 348.Nam YJ, Mani K, Wu L, Peng CF, Calvert JW, Foo RS, Krishnamurthy B, Miao W, Ashton AW, Lefer DJ, Kitsis RN. The apoptosis inhibitor ARC undergoes ubiquitin-proteasomal-mediated degradation in response to death stimuli: identification of a degradation-resistant mutant. J Biol Chem 282: 5522–5528, 2007. doi: 10.1074/jbc.M609186200. [DOI] [PubMed] [Google Scholar]
- 349.Naon D, Zaninello M, Giacomello M, Varanita T, Grespi F, Lakshminaranayan S, Serafini A, Semenzato M, Herkenne S, Hernández-Alvarez MI, Zorzano A, De Stefani D, Dorn GW II, Scorrano L. Critical reappraisal confirms that Mitofusin 2 is an endoplasmic reticulum-mitochondria tether. Proc Natl Acad Sci USA 113: 11249–11254, 2016. doi: 10.1073/pnas.1606786113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 350.Nazareth W, Yafei N, Crompton M. Inhibition of anoxia-induced injury in heart myocytes by cyclosporin A. J Mol Cell Cardiol 23: 1351–1354, 1991. doi: 10.1016/0022-2828(91)90181-K. [DOI] [PubMed] [Google Scholar]
- 351.Newton K, Dugger DL, Maltzman A, Greve JM, Hedehus M, Martin-McNulty B, Carano RA, Cao TC, van Bruggen N, Bernstein L, Lee WP, Wu X, DeVoss J, Zhang J, Jeet S, Peng I, McKenzie BS, Roose-Girma M, Caplazi P, Diehl L, Webster JD, Vucic D. RIPK3 deficiency or catalytically inactive RIPK1 provides greater benefit than MLKL deficiency in mouse models of inflammation and tissue injury. Cell Death Differ 23: 1565–1576, 2016. doi: 10.1038/cdd.2016.46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 352.Newton K, Dugger DL, Wickliffe KE, Kapoor N, de Almagro MC, Vucic D, Komuves L, Ferrando RE, French DM, Webster J, Roose-Girma M, Warming S, Dixit VM. Activity of protein kinase RIPK3 determines whether cells die by necroptosis or apoptosis. Science 343: 1357–1360, 2014. doi: 10.1126/science.1249361. [DOI] [PubMed] [Google Scholar]
- 353.Newton K, Manning G. Necroptosis and Inflammation. Annu Rev Biochem 85: 743–763, 2016. doi: 10.1146/annurev-biochem-060815-014830. [DOI] [PubMed] [Google Scholar]
- 354.Newton K, Sun X, Dixit VM. Kinase RIP3 is dispensable for normal NF-kappa Bs, signaling by the B-cell and T-cell receptors, tumor necrosis factor receptor 1, and Toll-like receptors 2 and 4. Mol Cell Biol 24: 1464–1469, 2004. doi: 10.1128/MCB.24.4.1464-1469.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 355.Nicholson DW, Thornberry NA. Caspases: killer proteases. Trends Biochem Sci 22: 299–306, 1997. doi: 10.1016/S0968-0004(97)01085-2. [DOI] [PubMed] [Google Scholar]
- 356.Nishikawa T, Ishiyama S, Nagata M, Sakomura Y, Nakazawa M, Momma K, Hiroe M, Kasajima T. Programmed cell death in the myocardium of arrhythmogenic right ventricular cardiomyopathy in children and adults. Cardiovasc Pathol 8: 185–189, 1999. doi: 10.1016/S1054-8807(99)00007-1. [DOI] [PubMed] [Google Scholar]
- 357.Nishimura H, Okazaki T, Tanaka Y, Nakatani K, Hara M, Matsumori A, Sasayama S, Mizoguchi A, Hiai H, Minato N, Honjo T. Autoimmune dilated cardiomyopathy in PD-1 receptor-deficient mice. Science 291: 319–322, 2001. doi: 10.1126/science.291.5502.319. [DOI] [PubMed] [Google Scholar]
- 358.Nishino M, Sholl LM, Hatabu H, Ramaiya NH, Hodi FS. Anti-PD-1-Related Pneumonitis during Cancer Immunotherapy. N Engl J Med 373: 288–290, 2015. doi: 10.1056/NEJMc1505197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 359.Oakes SA, Opferman JT, Pozzan T, Korsmeyer SJ, Scorrano L. Regulation of endoplasmic reticulum Ca2+ dynamics by proapoptotic BCL-2 family members. Biochem Pharmacol 66: 1335–1340, 2003. doi: 10.1016/S0006-2952(03)00482-9. [DOI] [PubMed] [Google Scholar]
- 360.Oakes SA, Scorrano L, Opferman JT, Bassik MC, Nishino M, Pozzan T, Korsmeyer SJ. Proapoptotic BAX and BAK regulate the type 1 inositol trisphosphate receptor and calcium leak from the endoplasmic reticulum. Proc Natl Acad Sci USA 102: 105–110, 2005. doi: 10.1073/pnas.0408352102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 361.Oberst A, Dillon CP, Weinlich R, McCormick LL, Fitzgerald P, Pop C, Hakem R, Salvesen GS, Green DR. Catalytic activity of the caspase-8-FLIP(L) complex inhibits RIPK3-dependent necrosis. Nature 471: 363–367, 2011. doi: 10.1038/nature09852. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 362.Oberst A, Pop C, Tremblay AG, Blais V, Denault JB, Salvesen GS, Green DR. Inducible dimerization and inducible cleavage reveal a requirement for both processes in caspase-8 activation. J Biol Chem 285: 16632–16642, 2010. doi: 10.1074/jbc.M109.095083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 363.Oberstein A, Jeffrey PD, Shi Y. Crystal structure of the Bcl-XL-Beclin 1 peptide complex: Beclin 1 is a novel BH3-only protein. J Biol Chem 282: 13123–13132, 2007. doi: 10.1074/jbc.M700492200. [DOI] [PubMed] [Google Scholar]
- 364.Oda E, Ohki R, Murasawa H, Nemoto J, Shibue T, Yamashita T, Tokino T, Taniguchi T, Tanaka N. Noxa, a BH3-only member of the Bcl-2 family and candidate mediator of p53-induced apoptosis. Science 288: 1053–1058, 2000. doi: 10.1126/science.288.5468.1053. [DOI] [PubMed] [Google Scholar]
- 365.Okazaki T, Tanaka Y, Nishio R, Mitsuiye T, Mizoguchi A, Wang J, Ishida M, Hiai H, Matsumori A, Minato N, Honjo T. Autoantibodies against cardiac troponin I are responsible for dilated cardiomyopathy in PD-1-deficient mice. Nat Med 9: 1477–1483, 2003. doi: 10.1038/nm955. [DOI] [PubMed] [Google Scholar]
- 366.Olivetti G, Abbi R, Quaini F, Kajstura J, Cheng W, Nitahara JA, Quaini E, Di Loreto C, Beltrami CA, Krajewski S, Reed JC, Anversa P. Apoptosis in the failing human heart. N Engl J Med 336: 1131–1141, 1997. doi: 10.1056/NEJM199704173361603. [DOI] [PubMed] [Google Scholar]
- 367.Ong SB, Samangouei P, Kalkhoran SB, Hausenloy DJ. The mitochondrial permeability transition pore and its role in myocardial ischemia reperfusion injury. J Mol Cell Cardiol 78: 23–34, 2015. doi: 10.1016/j.yjmcc.2014.11.005. [DOI] [PubMed] [Google Scholar]
- 368.Ong SB, Subrayan S, Lim SY, Yellon DM, Davidson SM, Hausenloy DJ. Inhibiting mitochondrial fission protects the heart against ischemia/reperfusion injury. Circulation 121: 2012–2022, 2010. doi: 10.1161/CIRCULATIONAHA.109.906610. [DOI] [PubMed] [Google Scholar]
- 369.Orlinick JR, Vaishnaw A, Elkon KB, Chao MV. Requirement of cysteine-rich repeats of the Fas receptor for binding by the Fas ligand. J Biol Chem 272: 28889–28894, 1997. doi: 10.1074/jbc.272.46.28889. [DOI] [PubMed] [Google Scholar]
- 370.Orning P, Weng D, Starheim K, Ratner D, Best Z, Lee B, Brooks A, Xia S, Wu H, Kelliher MA, Berger SB, Gough PJ, Bertin J, Proulx MM, Goguen JD, Kayagaki N, Fitzgerald KA, Lien E. Pathogen blockade of TAK1 triggers caspase-8-dependent cleavage of gasdermin D and cell death. Science 362: 1064–1069, 2018. doi: 10.1126/science.aau2818. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 371.Overholtzer M, Mailleux AA, Mouneimne G, Normand G, Schnitt SJ, King RW, Cibas ES, Brugge JS. A nonapoptotic cell death process, entosis, that occurs by cell-in-cell invasion. Cell 131: 966–979, 2007. doi: 10.1016/j.cell.2007.10.040. [DOI] [PubMed] [Google Scholar]
- 372.Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer 12: 252–264, 2012. doi: 10.1038/nrc3239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 373.Park HH, Lo YC, Lin SC, Wang L, Yang JK, Wu H. The death domain superfamily in intracellular signaling of apoptosis and inflammation. Annu Rev Immunol 25: 561–586, 2007. doi: 10.1146/annurev.immunol.25.022106.141656. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 374.Park S, Juliana C, Hong S, Datta P, Hwang I, Fernandes-Alnemri T, Yu JW, Alnemri ES. The mitochondrial antiviral protein MAVS associates with NLRP3 and regulates its inflammasome activity. J Immunol 191: 4358–4366, 2013. doi: 10.4049/jimmunol.1301170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 375.Patron M, Raffaello A, Granatiero V, Tosatto A, Merli G, De Stefani D, Wright L, Pallafacchina G, Terrin A, Mammucari C, Rizzuto R. The mitochondrial calcium uniporter (MCU): molecular identity and physiological roles. J Biol Chem 288: 10750–10758, 2013. doi: 10.1074/jbc.R112.420752. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 376.Patten IS, Rana S, Shahul S, Rowe GC, Jang C, Liu L, Hacker MR, Rhee JS, Mitchell J, Mahmood F, Hess P, Farrell C, Koulisis N, Khankin EV, Burke SD, Tudorache I, Bauersachs J, del Monte F, Hilfiker-Kleiner D, Karumanchi SA, Arany Z. Cardiac angiogenic imbalance leads to peripartum cardiomyopathy. Nature 485: 333–338, 2012. doi: 10.1038/nature11040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 377.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. [DOI] [PubMed] [Google Scholar]
- 378.Pieper AA, Walles T, Wei G, Clements EE, Verma A, Snyder SH, Zweier JL. Myocardial postischemic injury is reduced by polyADPribose polymerase-1 gene disruption. Mol Med 6: 271–282, 2000. doi: 10.1007/BF03401936. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 379.Pillai JB, Russell HM, Raman J, Jeevanandam V, Gupta MP. Increased expression of poly(ADP-ribose) polymerase-1 contributes to caspase-independent myocyte cell death during heart failure. Am J Physiol Heart Circ Physiol 288: H486–H496, 2005. doi: 10.1152/ajpheart.00437.2004. [DOI] [PubMed] [Google Scholar]
- 380.Pinkoski MJ, Waterhouse NJ, Heibein JA, Wolf BB, Kuwana T, Goldstein JC, Newmeyer DD, Bleackley RC, Green DR. Granzyme B-mediated apoptosis proceeds predominantly through a Bcl-2-inhibitable mitochondrial pathway. J Biol Chem 276: 12060–12067, 2001. doi: 10.1074/jbc.M009038200. [DOI] [PubMed] [Google Scholar]
- 381.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. [DOI] [PubMed] [Google Scholar]
- 382.Polykratis A, Hermance N, Zelic M, Roderick J, Kim C, Van TM, Lee TH, Chan FKM, Pasparakis M, Kelliher MA. Cutting edge: RIPK1 kinase inactive mice are viable and protected from TNF-induced necroptosis in vivo. J Immunol 193: 1539–1543, 2014. doi: 10.4049/jimmunol.1400590. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 383.Posadas I, Pérez-Martínez FC, Guerra J, Sánchez-Verdú P, Ceña V. Cofilin activation mediates Bax translocation to mitochondria during excitotoxic neuronal death. J Neurochem 120: 515–527, 2012. doi: 10.1111/j.1471-4159.2011.07599.x. [DOI] [PubMed] [Google Scholar]
- 384.Puthalakath H, O’Reilly LA, Gunn P, Lee L, Kelly PN, Huntington ND, Hughes PD, Michalak EM, McKimm-Breschkin J, Motoyama N, Gotoh T, Akira S, Bouillet P, Strasser A. ER stress triggers apoptosis by activating BH3-only protein Bim. Cell 129: 1337–1349, 2007. doi: 10.1016/j.cell.2007.04.027. [DOI] [PubMed] [Google Scholar]
- 385.Py BF, Kim MS, Vakifahmetoglu-Norberg H, Yuan J. Deubiquitination of NLRP3 by BRCC3 critically regulates inflammasome activity. Mol Cell 49: 331–338, 2013. doi: 10.1016/j.molcel.2012.11.009. [DOI] [PubMed] [Google Scholar]
- 386.Pyo JO, Nah J, Kim HJ, Chang JW, Song YW, Yang DK, Jo DG, Kim HR, Chae HJ, Chae SW, Hwang SY, Kim SJ, Kim HJ, Cho C, Oh CG, Park WJ, Jung YK. Protection of cardiomyocytes from ischemic/hypoxic cell death via Drbp1 and pMe2GlyDH in cardio-specific ARC transgenic mice. J Biol Chem 283: 30707–30714, 2008. doi: 10.1074/jbc.M804209200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 387.Qi D, Hu X, Wu X, Merk M, Leng L, Bucala R, Young LH. Cardiac macrophage migration inhibitory factor inhibits JNK pathway activation and injury during ischemia/reperfusion. J Clin Invest 119: 3807–3816, 2009. doi: 10.1172/JCI39738. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 388.Qin D, Wang X, Li Y, Yang L, Wang R, Peng J, Essandoh K, Mu X, Peng T, Han Q, Yu KJ, Fan GC. MicroRNA-223-5p and -3p Cooperatively Suppress Necroptosis in Ischemic/Reperfused Hearts. J Biol Chem 291: 20247–20259, 2016. doi: 10.1074/jbc.M116.732735. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 389.Raaflaub J. [Swelling of isolated mitochondria of the liver and their susceptibility to physicochemical influences]. Helv Physiol Pharmacol Acta 11: 142–156, 1953. [PubMed] [Google Scholar]
- 390.Ramirez MLG, Salvesen GS. A primer on caspase mechanisms. Semin Cell Dev Biol 82: 79–85, 2018. doi: 10.1016/j.semcdb.2018.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 391.Rathinam VA, Fitzgerald KA. Inflammasome Complexes: Emerging Mechanisms and Effector Functions. Cell 165: 792–800, 2016. doi: 10.1016/j.cell.2016.03.046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 392.Rathinam VA, Vanaja SK, Fitzgerald KA. Regulation of inflammasome signaling. Nat Immunol 13: 333–342, 2012. doi: 10.1038/ni.2237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 393.Rathinam VA, Vanaja SK, Waggoner L, Sokolovska A, Becker C, Stuart LM, Leong JM, Fitzgerald KA. TRIF licenses caspase-11-dependent NLRP3 inflammasome activation by gram-negative bacteria. Cell 150: 606–619, 2012. doi: 10.1016/j.cell.2012.07.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 394.Ray CA, Pickup DJ. The mode of death of pig kidney cells infected with cowpox virus is governed by the expression of the crmA gene. Virology 217: 384–391, 1996. doi: 10.1006/viro.1996.0128. [DOI] [PubMed] [Google Scholar]
- 395.Rebsamen M, Heinz LX, Meylan E, Michallet MC, Schroder K, Hofmann K, Vazquez J, Benedict CA, Tschopp J. DAI/ZBP1 recruits RIP1 and RIP3 through RIP homotypic interaction motifs to activate NF-kappaB. EMBO Rep 10: 916–922, 2009. doi: 10.1038/embor.2009.109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 396.Regula KM, Ens K, Kirshenbaum LA. Inducible expression of BNIP3 provokes mitochondrial defects and hypoxia-mediated cell death of ventricular myocytes. Circ Res 91: 226–231, 2002. doi: 10.1161/01.RES.0000029232.42227.16. [DOI] [PubMed] [Google Scholar]
- 397.Ren D, Tu HC, Kim H, Wang GX, Bean GR, Takeuchi O, Jeffers JR, Zambetti GP, Hsieh JJ, Cheng EH. BID, BIM, and PUMA are essential for activation of the BAX- and BAK-dependent cell death program. Science 330: 1390–1393, 2010. doi: 10.1126/science.1190217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 398.Reynoso E, Liu H, Li L, Yuan AL, Chen S, Wang Z. Thioredoxin-1 actively maintains the pseudokinase MLKL in a reduced state to suppress disulfide bond-dependent MLKL polymer formation and necroptosis. J Biol Chem 292: 17514–17524, 2017. doi: 10.1074/jbc.M117.799353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 399.Ribas A. Adaptive Immune Resistance: How Cancer Protects from Immune Attack. Cancer Discov 5: 915–919, 2015. doi: 10.1158/2159-8290.CD-15-0563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 400.Ribas A, Wolchok JD. Cancer immunotherapy using checkpoint blockade. Science 359: 1350–1355, 2018. doi: 10.1126/science.aar4060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 401.Ricci JE, Muñoz-Pinedo C, Fitzgerald P, Bailly-Maitre B, Perkins GA, Yadava N, Scheffler IE, Ellisman MH, Green DR. Disruption of mitochondrial function during apoptosis is mediated by caspase cleavage of the p75 subunit of complex I of the electron transport chain. Cell 117: 773–786, 2004. doi: 10.1016/j.cell.2004.05.008. [DOI] [PubMed] [Google Scholar]
- 402.Rickard JA, O’Donnell JA, Evans JM, Lalaoui N, Poh AR, Rogers T, Vince JE, Lawlor KE, Ninnis RL, Anderton H, Hall C, Spall SK, Phesse TJ, Abud HE, Cengia LH, Corbin J, Mifsud S, Di Rago L, Metcalf D, Ernst M, Dewson G, Roberts AW, Alexander WS, Murphy JM, Ekert PG, Masters SL, Vaux DL, Croker BA, Gerlic M, Silke J. RIPK1 regulates RIPK3-MLKL-driven systemic inflammation and emergency hematopoiesis. Cell 157: 1175–1188, 2014. doi: 10.1016/j.cell.2014.04.019. [DOI] [PubMed] [Google Scholar]
- 403.Riedl SJ, Renatus M, Schwarzenbacher R, Zhou Q, Sun C, Fesik SW, Liddington RC, Salvesen GS. Structural basis for the inhibition of caspase-3 by XIAP. Cell 104: 791–800, 2001. doi: 10.1016/S0092-8674(01)00274-4. [DOI] [PubMed] [Google Scholar]
- 404.Robbins SL. Pathologic Basis of Disease. Philadelphia, PA: Saunders, 1974. [Google Scholar]
- 405.Roderick JE, Hermance N, Zelic M, Simmons MJ, Polykratis A, Pasparakis M, Kelliher MA. Hematopoietic RIPK1 deficiency results in bone marrow failure caused by apoptosis and RIPK3-mediated necroptosis. Proc Natl Acad Sci USA 111: 14436–14441, 2014. doi: 10.1073/pnas.1409389111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 406.Rogers C, Fernandes-Alnemri T, Mayes L, Alnemri D, Cingolani G, Alnemri ES. Cleavage of DFNA5 by caspase-3 during apoptosis mediates progression to secondary necrotic/pyroptotic cell death. Nat Commun 8: 14128, 2017. doi: 10.1038/ncomms14128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 407.Roy N, Deveraux QL, Takahashi R, Salvesen GS, Reed JC. The c-IAP-1 and c-IAP-2 proteins are direct inhibitors of specific caspases. EMBO J 16: 6914–6925, 1997. doi: 10.1093/emboj/16.23.6914. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 409.Rück A, Dolder M, Wallimann T, Brdiczka D. Reconstituted adenine nucleotide translocase forms a channel for small molecules comparable to the mitochondrial permeability transition pore. FEBS Lett 426: 97–101, 1998. doi: 10.1016/S0014-5793(98)00317-2. [DOI] [PubMed] [Google Scholar]
- 410.Saelens X, Festjens N, Parthoens E, Vanoverberghe I, Kalai M, van Kuppeveld F, Vandenabeele P. Protein synthesis persists during necrotic cell death. J Cell Biol 168: 545–551, 2005. doi: 10.1083/jcb.200407162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 411.Sahara S, Aoto M, Eguchi Y, Imamoto N, Yoneda Y, Tsujimoto Y. Acinus is a caspase-3-activated protein required for apoptotic chromatin condensation. Nature 401: 168–173, 1999. doi: 10.1038/43678. [DOI] [PubMed] [Google Scholar]
- 412.Sakahira H, Enari M, Nagata S. Cleavage of CAD inhibitor in CAD activation and DNA degradation during apoptosis. [Corrigendum in Nature 526: 728, 2015.] Nature 391: 96–99, 1998. doi: 10.1038/34214. [DOI] [PubMed] [Google Scholar]
- 413.Saleh M, Vaillancourt JP, Graham RK, Huyck M, Srinivasula SM, Alnemri ES, Steinberg MH, Nolan V, Baldwin CT, Hotchkiss RS, Buchman TG, Zehnbauer BA, Hayden MR, Farrer LA, Roy S, Nicholson DW. Differential modulation of endotoxin responsiveness by human caspase-12 polymorphisms. Nature 429: 75–79, 2004. doi: 10.1038/nature02451. [DOI] [PubMed] [Google Scholar]
- 414.Salem JE, Manouchehri A, Moey M, Lebrun-Vignes B, Bastarache L, Pariente A, Gobert A, Spano JP, Balko JM, Bonaca MP, Roden DM, Johnson DB, Moslehi JJ. Cardiovascular toxicities associated with immune checkpoint inhibitors: an observational, retrospective, pharmacovigilance study. Lancet Oncol 19: 1579–1589, 2018. doi: 10.1016/S1470-2045(18)30608-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 415.Sanchis D, Llovera M, Ballester M, Comella JX. An alternative view of apoptosis in heart development and disease. Cardiovasc Res 77: 448–451, 2008. doi: 10.1093/cvr/cvm074. [DOI] [PubMed] [Google Scholar]
- 416.Sanchis D, Mayorga M, Ballester M, Comella JX. Lack of Apaf-1 expression confers resistance to cytochrome c-driven apoptosis in cardiomyocytes. Cell Death Differ 10: 977–986, 2003. doi: 10.1038/sj.cdd.4401267. [DOI] [PubMed] [Google Scholar]
- 417.Sandanger Ø, Ranheim T, Vinge LE, Bliksøen M, Alfsnes K, Finsen AV, Dahl CP, Askevold ET, Florholmen G, Christensen G, Fitzgerald KA, Lien E, Valen G, Espevik T, Aukrust P, Yndestad A. The NLRP3 inflammasome is up-regulated in cardiac fibroblasts and mediates myocardial ischaemia-reperfusion injury. Cardiovasc Res 99: 164–174, 2013. doi: 10.1093/cvr/cvt091. [DOI] [PubMed] [Google Scholar]
- 418.Saraste A, Pulkki K, Kallajoki M, Heikkilä P, Laine P, Mattila S, Nieminen MS, Parvinen M, Voipio-Pulkki LM. Cardiomyocyte apoptosis and progression of heart failure to transplantation. Eur J Clin Invest 29: 380–386, 1999. doi: 10.1046/j.1365-2362.1999.00481.x. [DOI] [PubMed] [Google Scholar]
- 419.Saraste A, Pulkki K, Kallajoki M, Henriksen K, Parvinen M, Voipio-Pulkki LM. Apoptosis in human acute myocardial infarction. Circulation 95: 320–323, 1997. doi: 10.1161/01.CIR.95.2.320. [DOI] [PubMed] [Google Scholar]
- 420.Saraste A, Voipio-Pulkki LM, Parvinen M, Pulkki K. Apoptosis in the heart. N Engl J Med 336: 1025–1026, 1997. doi: 10.1056/NEJM199704033361415. [DOI] [PubMed] [Google Scholar]
- 421.Sarosiek KA, Fraser C, Muthalagu N, Bhola PD, Chang W, McBrayer SK, Cantlon A, Fisch S, Golomb-Mello G, Ryan JA, Deng J, Jian B, Corbett C, Goldenberg M, Madsen JR, Liao R, Walsh D, Sedivy J, Murphy DJ, Carrasco DR, Robinson S, Moslehi J, Letai A. Developmental Regulation of Mitochondrial Apoptosis by c-Myc Governs Age- and Tissue-Specific Sensitivity to Cancer Therapeutics. Cancer Cell 31: 142–156, 2017. doi: 10.1016/j.ccell.2016.11.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 422.Saunders JW Jr, Gasseling MT, Saunders LC. Cellular death in morphogenesis of the avian wing. Dev Biol 5: 147–178, 1962. doi: 10.1016/0012-1606(62)90008-8. [DOI] [PubMed] [Google Scholar]
- 423.Scarabelli T, Stephanou A, Rayment N, Pasini E, Comini L, Curello S, Ferrari R, Knight R, Latchman D. Apoptosis of endothelial cells precedes myocyte cell apoptosis in ischemia/reperfusion injury. Circulation 104: 253–256, 2001. doi: 10.1161/01.CIR.104.3.253. [DOI] [PubMed] [Google Scholar]
- 424.Schile AJ, García-Fernández M, Steller H. Regulation of apoptosis by XIAP ubiquitin-ligase activity. Genes Dev 22: 2256–2266, 2008. doi: 10.1101/gad.1663108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 425.Schinzel AC, Takeuchi O, Huang Z, Fisher JK, Zhou Z, Rubens J, Hetz C, Danial NN, Moskowitz MA, Korsmeyer SJ. Cyclophilin D is a component of mitochondrial permeability transition and mediates neuronal cell death after focal cerebral ischemia. Proc Natl Acad Sci USA 102: 12005–12010, 2005. doi: 10.1073/pnas.0505294102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 426.Schlicher L, Wissler M, Preiss F, Brauns-Schubert P, Jakob C, Dumit V, Borner C, Dengjel J, Maurer U. SPATA2 promotes CYLD activity and regulates TNF-induced NF-κB signaling and cell death. EMBO Rep 17: 1485–1497, 2016. doi: 10.15252/embr.201642592. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 427.Sciarretta S, Zhai P, Shao D, Maejima Y, Robbins J, Volpe M, Condorelli G, Sadoshima J. Rheb is a critical regulator of autophagy during myocardial ischemia: pathophysiological implications in obesity and metabolic syndrome. Circulation 125: 1134–1146, 2012. doi: 10.1161/CIRCULATIONAHA.111.078212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 428.Sciarretta S, Zhai P, Shao D, Zablocki D, Nagarajan N, Terada LS, Volpe M, Sadoshima J. Activation of NADPH oxidase 4 in the endoplasmic reticulum promotes cardiomyocyte autophagy and survival during energy stress through the protein kinase RNA-activated-like endoplasmic reticulum kinase/eukaryotic initiation factor 2α/activating transcription factor 4 pathway. Circ Res 113: 1253–1264, 2013. doi: 10.1161/CIRCRESAHA.113.301787. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 429.Scorrano L, Ashiya M, Buttle K, Weiler S, Oakes SA, Mannella CA, Korsmeyer SJ. A distinct pathway remodels mitochondrial cristae and mobilizes cytochrome c during apoptosis. Dev Cell 2: 55–67, 2002. doi: 10.1016/S1534-5807(01)00116-2. [DOI] [PubMed] [Google Scholar]
- 430.Scorrano L, Oakes SA, Opferman JT, Cheng EH, Sorcinelli MD, Pozzan T, Korsmeyer SJ. BAX and BAK regulation of endoplasmic reticulum Ca2+: a control point for apoptosis. Science 300: 135–139, 2003. doi: 10.1126/science.1081208. [DOI] [PubMed] [Google Scholar]
- 431.Scott FL, Stec B, Pop C, Dobaczewska MK, Lee JJ, Monosov E, Robinson H, Salvesen GS, Schwarzenbacher R, Riedl SJ. The Fas-FADD death domain complex structure unravels signalling by receptor clustering. Nature 457: 1019–1022, 2009. doi: 10.1038/nature07606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 432.Segawa K, Kurata S, Yanagihashi Y, Brummelkamp TR, Matsuda F, Nagata S. Caspase-mediated cleavage of phospholipid flippase for apoptotic phosphatidylserine exposure. Science 344: 1164–1168, 2014. doi: 10.1126/science.1252809. [DOI] [PubMed] [Google Scholar]
- 433.Serrano-Puebla A, Boya P. Lysosomal membrane permeabilization in cell death: new evidence and implications for health and disease. Ann N Y Acad Sci 1371: 30–44, 2016. doi: 10.1111/nyas.12966. [DOI] [PubMed] [Google Scholar]
- 434.Shan B, Pan H, Najafov A, Yuan J. Necroptosis in development and diseases. Genes Dev 32: 327–340, 2018. doi: 10.1101/gad.312561.118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 435.Shekhar A, Heeger P, Reutelingsperger C, Arbustini E, Narula N, Hofstra L, Bax JJ, Narula J. Targeted Imaging for Cell Death in Cardiovascular Disorders. JACC Cardiovasc Imaging 11: 476–493, 2018. doi: 10.1016/j.jcmg.2017.11.018. [DOI] [PubMed] [Google Scholar]
- 436.Sheridan C, Delivani P, Cullen SP, Martin SJ. Bax- or Bak-induced mitochondrial fission can be uncoupled from cytochrome c release. Mol Cell 31: 570–585, 2008. doi: 10.1016/j.molcel.2008.08.002. [DOI] [PubMed] [Google Scholar]
- 437.Shi CS, Shenderov K, Huang NN, Kabat J, Abu-Asab M, Fitzgerald KA, Sher A, Kehrl JH. Activation of autophagy by inflammatory signals limits IL-1β production by targeting ubiquitinated inflammasomes for destruction. Nat Immunol 13: 255–263, 2012. doi: 10.1038/ni.2215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 438.Shi J, Gao W, Shao F. Pyroptosis: Gasdermin-Mediated Programmed Necrotic Cell Death. Trends Biochem Sci 42: 245–254, 2017. doi: 10.1016/j.tibs.2016.10.004. [DOI] [PubMed] [Google Scholar]
- 439.Shi J, Zhao Y, Wang K, Shi X, Wang Y, Huang H, Zhuang Y, Cai T, Wang F, Shao F. Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nature 526: 660–665, 2015. doi: 10.1038/nature15514. [DOI] [PubMed] [Google Scholar]
- 440.Shi J, Zhao Y, Wang Y, Gao W, Ding J, Li P, Hu L, Shao F. Inflammatory caspases are innate immune receptors for intracellular LPS. Nature 514: 187–192, 2014. doi: 10.1038/nature13683. [DOI] [PubMed] [Google Scholar]
- 441.Shimada K, Crother TR, Karlin J, Dagvadorj J, Chiba N, Chen S, Ramanujan VK, Wolf AJ, Vergnes L, Ojcius DM, Rentsendorj A, Vargas M, Guerrero C, Wang Y, Fitzgerald KA, Underhill DM, Town T, Arditi M. Oxidized mitochondrial DNA activates the NLRP3 inflammasome during apoptosis. Immunity 36: 401–414, 2012. doi: 10.1016/j.immuni.2012.01.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 442.Shiozaki EN, Chai J, Rigotti DJ, Riedl SJ, Li P, Srinivasula SM, Alnemri ES, Fairman R, Shi Y. Mechanism of XIAP-mediated inhibition of caspase-9. Mol Cell 11: 519–527, 2003. doi: 10.1016/S1097-2765(03)00054-6. [DOI] [PubMed] [Google Scholar]
- 443.Shoshan-Barmatz V, De S, Meir A. The Mitochondrial Voltage-Dependent Anion Channel 1, Ca2+ Transport, Apoptosis, and Their Regulation. Front Oncol 7: 60, 2017. doi: 10.3389/fonc.2017.00060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 444.Shutinoski B, Alturki NA, Rijal D, Bertin J, Gough PJ, Schlossmacher MG, Sad S. K45A mutation of RIPK1 results in poor necroptosis and cytokine signaling in macrophages, which impacts inflammatory responses in vivo. Cell Death Differ 23: 1628–1637, 2016. doi: 10.1038/cdd.2016.51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 445.Siegel RM, Frederiksen JK, Zacharias DA, Chan FK, Johnson M, Lynch D, Tsien RY, Lenardo MJ. Fas preassociation required for apoptosis signaling and dominant inhibition by pathogenic mutations. Science 288: 2354–2357, 2000. doi: 10.1126/science.288.5475.2354. [DOI] [PubMed] [Google Scholar]
- 446.Skouta R, Dixon SJ, Wang J, Dunn DE, Orman M, Shimada K, Rosenberg PA, Lo DC, Weinberg JM, Linkermann A, Stockwell BR. Ferrostatins inhibit oxidative lipid damage and cell death in diverse disease models. J Am Chem Soc 136: 4551–4556, 2014. doi: 10.1021/ja411006a. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 447.Slee EA, Adrain C, Martin SJ. Executioner caspase-3, -6, and -7 perform distinct, non-redundant roles during the demolition phase of apoptosis. J Biol Chem 276: 7320–7326, 2001. doi: 10.1074/jbc.M008363200. [DOI] [PubMed] [Google Scholar]
- 448.Smith CC, Davidson SM, Lim SY, Simpkin JC, Hothersall JS, Yellon DM. Necrostatin: a potentially novel cardioprotective agent? Cardiovasc Drugs Ther 21: 227–233, 2007. doi: 10.1007/s10557-007-6035-1. [DOI] [PubMed] [Google Scholar]
- 449.Soldani C, Scovassi AI. Poly(ADP-ribose) polymerase-1 cleavage during apoptosis: an update. Apoptosis 7: 321–328, 2002. doi: 10.1023/A:1016119328968. [DOI] [PubMed] [Google Scholar]
- 450.Song H, Liu B, Huai W, Yu Z, Wang W, Zhao J, Han L, Jiang G, Zhang L, Gao C, Zhao W. The E3 ubiquitin ligase TRIM31 attenuates NLRP3 inflammasome activation by promoting proteasomal degradation of NLRP3. Nat Commun 7: 13727, 2016. doi: 10.1038/ncomms13727. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 451.Song N, Liu ZS, Xue W, Bai ZF, Wang QY, Dai J, Liu X, Huang YJ, Cai H, Zhan XY, Han QY, Wang H, Chen Y, Li HY, Li AL, Zhang XM, Zhou T, Li T. NLRP3 Phosphorylation Is an Essential Priming Event for Inflammasome Activation. Mol Cell 68: 185–197.e6, 2017. doi: 10.1016/j.molcel.2017.08.017. [DOI] [PubMed] [Google Scholar]
- 452.Spalinger MR, Kasper S, Gottier C, Lang S, Atrott K, Vavricka SR, Scharl S, Raselli T, Frey-Wagner I, Gutte PM, Grütter MG, Beer HD, Contassot E, Chan AC, Dai X, Rawlings DJ, Mair F, Becher B, Falk W, Fried M, Rogler G, Scharl M. NLRP3 tyrosine phosphorylation is controlled by protein tyrosine phosphatase PTPN22. J Clin Invest 126: 4388, 2016. doi: 10.1172/JCI90897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 453.Srinivasula SM, Hegde R, Saleh A, Datta P, Shiozaki E, Chai J, Lee RA, Robbins PD, Fernandes-Alnemri T, Shi Y, Alnemri ES. A conserved XIAP-interaction motif in caspase-9 and Smac/DIABLO regulates caspase activity and apoptosis. [Erratum in Nature 411: 1081, 2001.] Nature 410: 112–116, 2001. doi: 10.1038/35065125. [DOI] [PubMed] [Google Scholar]
- 454.Stanger BZ, Leder P, Lee TH, Kim E, Seed B. RIP: a novel protein containing a death domain that interacts with Fas/APO-1 (CD95) in yeast and causes cell death. Cell 81: 513–523, 1995. doi: 10.1016/0092-8674(95)90072-1. [DOI] [PubMed] [Google Scholar]
- 455.Starling GC, Bajorath J, Emswiler J, Ledbetter JA, Aruffo A, Kiener PA. Identification of amino acid residues important for ligand binding to Fas. J Exp Med 185: 1487–1492, 1997. doi: 10.1084/jem.185.8.1487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 456.Stockwell BR, Friedmann Angeli JP, Bayir H, Bush AI, Conrad M, Dixon SJ, Fulda S, Gascón S, Hatzios SK, Kagan VE, Noel K, Jiang X, Linkermann A, Murphy ME, Overholtzer M, Oyagi A, Pagnussat GC, Park J, Ran Q, Rosenfeld CS, Salnikow K, Tang D, Torti FM, Torti SV, Toyokuni S, Woerpel KA, Zhang DD. Ferroptosis: A Regulated Cell Death Nexus Linking Metabolism, Redox Biology, and Disease. Cell 171: 273–285, 2017. doi: 10.1016/j.cell.2017.09.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 457.Stutz A, Kolbe CC, Stahl R, Horvath GL, Franklin BS, van Ray O, Brinkschulte R, Geyer M, Meissner F, Latz E. NLRP3 inflammasome assembly is regulated by phosphorylation of the pyrin domain. J Exp Med 214: 1725–1736, 2017. doi: 10.1084/jem.20160933. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 458.Subramanian N, Natarajan K, Clatworthy MR, Wang Z, Germain RN. The adaptor MAVS promotes NLRP3 mitochondrial localization and inflammasome activation. Cell 153: 348–361, 2013. doi: 10.1016/j.cell.2013.02.054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 459.Suetomi T, Willeford A, Brand CS, Cho Y, Ross RS, Miyamoto S, Brown JH. Inflammation and NLRP3 Inflammasome Activation Initiated in Response to Pressure Overload by Ca2+/Calmodulin-Dependent Protein Kinase II δ Signaling in Cardiomyocytes Are Essential for Adverse Cardiac Remodeling. Circulation 138: 2530–2544, 2018. doi: 10.1161/CIRCULATIONAHA.118.034621. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 460.Sun L, Wang H, Wang Z, He S, Chen S, Liao D, Wang L, Yan J, Liu W, Lei X, Wang X. Mixed lineage kinase domain-like protein mediates necrosis signaling downstream of RIP3 kinase. Cell 148: 213–227, 2012. doi: 10.1016/j.cell.2011.11.031. [DOI] [PubMed] [Google Scholar]
- 461.Sun XM, Butterworth M, MacFarlane M, Dubiel W, Ciechanover A, Cohen GM. Caspase activation inhibits proteasome function during apoptosis. Mol Cell 14: 81–93, 2004. doi: 10.1016/S1097-2765(04)00156-X. [DOI] [PubMed] [Google Scholar]
- 462.Susin SA, Lorenzo HK, Zamzami N, Marzo I, Snow BE, Brothers GM, Mangion J, Jacotot E, Costantini P, Loeffler M, Larochette N, Goodlett DR, Aebersold R, Siderovski DP, Penninger JM, Kroemer G. Molecular characterization of mitochondrial apoptosis-inducing factor. Nature 397: 441–446, 1999. doi: 10.1038/17135. [DOI] [PubMed] [Google Scholar]
- 463.Suzuki J, Denning DP, Imanishi E, Horvitz HR, Nagata S. Xk-related protein 8 and CED-8 promote phosphatidylserine exposure in apoptotic cells. Science 341: 403–406, 2013. doi: 10.1126/science.1236758. [DOI] [PubMed] [Google Scholar]
- 464.Suzuki Y, Imai Y, Nakayama H, Takahashi K, Takio K, Takahashi R. A serine protease, HtrA2, is released from the mitochondria and interacts with XIAP, inducing cell death. Mol Cell 8: 613–621, 2001. doi: 10.1016/S1097-2765(01)00341-0. [DOI] [PubMed] [Google Scholar]
- 465.Suzuki Y, Nakabayashi Y, Nakata K, Reed JC, Takahashi R. X-linked inhibitor of apoptosis protein (XIAP) inhibits caspase-3 and -7 in distinct modes. J Biol Chem 276: 27058–27063, 2001. doi: 10.1074/jbc.M102415200. [DOI] [PubMed] [Google Scholar]
- 466.Suzuki Y, Nakabayashi Y, Takahashi R. Ubiquitin-protein ligase activity of X-linked inhibitor of apoptosis protein promotes proteasomal degradation of caspase-3 and enhances its anti-apoptotic effect in Fas-induced cell death. Proc Natl Acad Sci USA 98: 8662–8667, 2001. doi: 10.1073/pnas.161506698. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 467.Syed FM, Hahn HS, Odley A, Guo Y, Vallejo JG, Lynch RA, Mann DL, Bolli R, Dorn GW II. Proapoptotic effects of caspase-1/interleukin-converting enzyme dominate in myocardial ischemia. Circ Res 96: 1103–1109, 2005. doi: 10.1161/01.RES.0000166925.45995.ed. [DOI] [PubMed] [Google Scholar]
- 468.Tait SW, Oberst A, Quarato G, Milasta S, Haller M, Wang R, Karvela M, Ichim G, Yatim N, Albert ML, Kidd G, Wakefield R, Frase S, Krautwald S, Linkermann A, Green DR. Widespread mitochondrial depletion via mitophagy does not compromise necroptosis. Cell Rep 5: 878–885, 2013. doi: 10.1016/j.celrep.2013.10.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 469.Takahashi K, Kawai T, Kumar H, Sato S, Yonehara S, Akira S. Roles of caspase-8 and caspase-10 in innate immune responses to double-stranded RNA. J Immunol 176: 4520–4524, 2006. doi: 10.4049/jimmunol.176.8.4520. [DOI] [PubMed] [Google Scholar]
- 470.Takahashi N, Duprez L, Grootjans S, Cauwels A, Nerinckx W, DuHadaway JB, Goossens V, Roelandt R, Van Hauwermeiren F, Libert C, Declercq W, Callewaert N, Prendergast GC, Degterev A, Yuan J, Vandenabeele P. Necrostatin-1 analogues: critical issues on the specificity, activity and in vivo use in experimental disease models. Cell Death Dis 3: e437, 2012. doi: 10.1038/cddis.2012.176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 471.Takahashi N, Vereecke L, Bertrand MJ, Duprez L, Berger SB, Divert T, Gonçalves A, Sze M, Gilbert B, Kourula S, Goossens V, Lefebvre S, Günther C, Becker C, Bertin J, Gough PJ, Declercq W, van Loo G, Vandenabeele P. RIPK1 ensures intestinal homeostasis by protecting the epithelium against apoptosis. Nature 513: 95–99, 2014. doi: 10.1038/nature13706. [DOI] [PubMed] [Google Scholar]
- 472.Takemura G, Fujiwara H. Role of apoptosis in remodeling after myocardial infarction. Pharmacol Ther 104: 1–16, 2004. doi: 10.1016/j.pharmthera.2004.07.005. [DOI] [PubMed] [Google Scholar]
- 473.Takemura G, Ohno M, Hayakawa Y, Misao J, Kanoh M, Ohno A, Uno Y, Minatoguchi S, Fujiwara T, Fujiwara H. Role of apoptosis in the disappearance of infiltrated and proliferated interstitial cells after myocardial infarction. Circ Res 82: 1130–1138, 1998. doi: 10.1161/01.RES.82.11.1130. [DOI] [PubMed] [Google Scholar]
- 474.Tang D, Kang R, Coyne CB, Zeh HJ, Lotze MT. PAMPs and DAMPs: signal 0s that spur autophagy and immunity. Immunol Rev 249: 158–175, 2012. doi: 10.1111/j.1600-065X.2012.01146.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 475.Tannous P, Zhu H, Johnstone JL, Shelton JM, Rajasekaran NS, Benjamin IJ, Nguyen L, Gerard RD, Levine B, Rothermel BA, Hill JA. Autophagy is an adaptive response in desmin-related cardiomyopathy. Proc Natl Acad Sci USA 105: 9745–9750, 2008. doi: 10.1073/pnas.0706802105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 476.Tarrio ML, Grabie N, Bu DX, Sharpe AH, Lichtman AH. PD-1 protects against inflammation and myocyte damage in T cell-mediated myocarditis. J Immunol 188: 4876–4884, 2012. doi: 10.4049/jimmunol.1200389. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 477.Tenev T, Bianchi K, Darding M, Broemer M, Langlais C, Wallberg F, Zachariou A, Lopez J, MacFarlane M, Cain K, Meier P. The Ripoptosome, a signaling platform that assembles in response to genotoxic stress and loss of IAPs. Mol Cell 43: 432–448, 2011. doi: 10.1016/j.molcel.2011.06.006. [DOI] [PubMed] [Google Scholar]
- 478.Théroux P, Chaitman BR, Danchin N, Erhardt L, Meinertz T, Schroeder JS, Tognoni G, White HD, Willerson JT, Jessel A. Inhibition of the sodium-hydrogen exchanger with cariporide to prevent myocardial infarction in high-risk ischemic situations. Main results of the GUARDIAN trial. Guard during ischemia against necrosis (GUARDIAN) Investigators. Circulation 102: 3032–3038, 2000. doi: 10.1161/01.CIR.102.25.3032. [DOI] [PubMed] [Google Scholar]
- 479.Thomas DA, Scorrano L, Putcha GV, Korsmeyer SJ, Ley TJ. Granzyme B can cause mitochondrial depolarization and cell death in the absence of BID, BAX, and BAK. Proc Natl Acad Sci USA 98: 14985–14990, 2001. doi: 10.1073/pnas.261581498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 480.Thornberry NA, Rano TA, Peterson EP, Rasper DM, Timkey T, Garcia-Calvo M, Houtzager VM, Nordstrom PA, Roy S, Vaillancourt JP, Chapman KT, Nicholson DW. A combinatorial approach defines specificities of members of the caspase family and granzyme B. Functional relationships established for key mediators of apoptosis. J Biol Chem 272: 17907–17911, 1997. doi: 10.1074/jbc.272.29.17907. [DOI] [PubMed] [Google Scholar]
- 481.Tivol EA, Borriello F, Schweitzer AN, Lynch WP, Bluestone JA, Sharpe AH. Loss of CTLA-4 leads to massive lymphoproliferation and fatal multiorgan tissue destruction, revealing a critical negative regulatory role of CTLA-4. Immunity 3: 541–547, 1995. doi: 10.1016/1074-7613(95)90125-6. [DOI] [PubMed] [Google Scholar]
- 482.Tonnus W, Gembardt F, Hugo C, Linkermann A. Die later with ESCRT! Oncotarget 8: 41790–41791, 2017. doi: 10.18632/oncotarget.17903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 483.Toth A, Jeffers JR, Nickson P, Min JY, Morgan JP, Zambetti GP, Erhardt P. Targeted deletion of Puma attenuates cardiomyocyte death and improves cardiac function during ischemia-reperfusion. Am J Physiol Heart Circ Physiol 291: H52–H60, 2006. doi: 10.1152/ajpheart.01046.2005. [DOI] [PubMed] [Google Scholar]
- 484.Tóth-Zsámboki E, Horváth E, Vargova K, Pankotai E, Murthy K, Zsengellér Z, Bárány T, Pék T, Fekete K, Kiss RG, Préda I, Lacza Z, Gerö D, Szabó C. Activation of poly(ADP-ribose) polymerase by myocardial ischemia and coronary reperfusion in human circulating leukocytes. Mol Med 12: 221–228, 2006. doi: 10.2119/2006-00055.Toth-Zsamboki. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 485.Trompouki E, Hatzivassiliou E, Tsichritzis T, Farmer H, Ashworth A, Mosialos G. CYLD is a deubiquitinating enzyme that negatively regulates NF-kappaB activation by TNFR family members. Nature 424: 793–796, 2003. doi: 10.1038/nature01803. [DOI] [PubMed] [Google Scholar]
- 486.Ucker DS. Exploiting death: apoptotic immunity in microbial pathogenesis. Cell Death Differ 23: 990–996, 2016. doi: 10.1038/cdd.2016.17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 487.Upton JW, Kaiser WJ, Mocarski ES. DAI/ZBP1/DLM-1 complexes with RIP3 to mediate virus-induced programmed necrosis that is targeted by murine cytomegalovirus vIRA. Cell Host Microbe 11: 290–297, 2012. doi: 10.1016/j.chom.2012.01.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 488.Upton JW, Kaiser WJ, Mocarski ES. Virus inhibition of RIP3-dependent necrosis. Cell Host Microbe 7: 302–313, 2010. doi: 10.1016/j.chom.2010.03.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 489.Vajen T, Koenen RR, Werner I, Staudt M, Projahn D, Curaj A, Sönmez TT, Simsekyilmaz S, Schumacher D, Möllmann J, Hackeng TM, Hundelshausen PV, Weber C, Liehn EA. Blocking CCL5-CXCL4 heteromerization preserves heart function after myocardial infarction by attenuating leukocyte recruitment and NETosis. Sci Rep 8: 10647, 2018. doi: 10.1038/s41598-018-29026-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 490.Van TM, Polykratis A, Straub BK, Kondylis V, Papadopoulou N, Pasparakis M. Kinase-independent functions of RIPK1 regulate hepatocyte survival and liver carcinogenesis. J Clin Invest 127: 2662–2677, 2017. doi: 10.1172/JCI92508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 491.Vande Walle L, Van Opdenbosch N, Jacques P, Fossoul A, Verheugen E, Vogel P, Beyaert R, Elewaut D, Kanneganti TD, van Loo G, Lamkanfi M. Negative regulation of the NLRP3 inflammasome by A20 protects against arthritis. Nature 512: 69–73, 2014. doi: 10.1038/nature13322. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 492.Vanden Berghe T, Linkermann A, Jouan-Lanhouet S, Walczak H, Vandenabeele P. Regulated necrosis: the expanding network of non-apoptotic cell death pathways. Nat Rev Mol Cell Biol 15: 135–147, 2014. doi: 10.1038/nrm3737. [DOI] [PubMed] [Google Scholar]
- 493.Van der Merwe PA, Bodian DL, Daenke S, Linsley P, Davis SJ. CD80 (B7-1) binds both CD28 and CTLA-4 with a low affinity and very fast kinetics. J Exp Med 185: 393–403, 1997. doi: 10.1084/jem.185.3.393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 494.Varfolomeev E, Goncharov T, Fedorova AV, Dynek JN, Zobel K, Deshayes K, Fairbrother WJ, Vucic D. c-IAP1 and c-IAP2 are critical mediators of tumor necrosis factor alpha (TNFalpha)-induced NF-kappaB activation. J Biol Chem 283: 24295–24299, 2008. doi: 10.1074/jbc.C800128200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 495.Varfolomeev EE, Schuchmann M, Luria V, Chiannilkulchai N, Beckmann JS, Mett IL, Rebrikov D, Brodianski VM, Kemper OC, Kollet O, Lapidot T, Soffer D, Sobe T, Avraham KB, Goncharov T, Holtmann H, Lonai P, Wallach D. Targeted disruption of the mouse Caspase 8 gene ablates cell death induction by the TNF receptors, Fas/Apo1, and DR3 and is lethal prenatally. Immunity 9: 267–276, 1998. doi: 10.1016/S1074-7613(00)80609-3. [DOI] [PubMed] [Google Scholar]
- 496.Vercammen D, Brouckaert G, Denecker G, Van de Craen M, Declercq W, Fiers W, Vandenabeele P. Dual signaling of the Fas receptor: initiation of both apoptotic and necrotic cell death pathways. J Exp Med 188: 919–930, 1998. doi: 10.1084/jem.188.5.919. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 497.Verhagen AM, Ekert PG, Pakusch M, Silke J, Connolly LM, Reid GE, Moritz RL, Simpson RJ, Vaux DL. Identification of DIABLO, a mammalian protein that promotes apoptosis by binding to and antagonizing IAP proteins. Cell 102: 43–53, 2000. doi: 10.1016/S0092-8674(00)00009-X. [DOI] [PubMed] [Google Scholar]
- 498.Vila MC, Rayavarapu S, Hogarth MW, Van der Meulen JH, Horn A, Defour A, Takeda S, Brown KJ, Hathout Y, Nagaraju K, Jaiswal JK. Mitochondria mediate cell membrane repair and contribute to Duchenne muscular dystrophy. Cell Death Differ 24: 330–342, 2017. doi: 10.1038/cdd.2016.127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 499.Virchow R. Cellular Pathology, translated by Chance F. New York: Dover, 1971. [Google Scholar]
- 500.Von Karstedt S, Montinaro A, Walczak H. Exploring the TRAILs less travelled: TRAIL in cancer biology and therapy. Nat Rev Cancer 17: 352–366, 2017. doi: 10.1038/nrc.2017.28. [DOI] [PubMed] [Google Scholar]
- 501.Voskoboinik I, Whisstock JC, Trapani JA. Perforin and granzymes: function, dysfunction and human pathology. Nat Rev Immunol 15: 388–400, 2015. doi: 10.1038/nri3839. [DOI] [PubMed] [Google Scholar]
- 502.Vucic D, Dixit VM, Wertz IE. Ubiquitylation in apoptosis: a post-translational modification at the edge of life and death. Nat Rev Mol Cell Biol 12: 439–452, 2011. doi: 10.1038/nrm3143. [DOI] [PubMed] [Google Scholar]
- 503.Wang H, Meng H, Li X, Zhu K, Dong K, Mookhtiar AK, Wei H, Li Y, Sun SC, Yuan J. PELI1 functions as a dual modulator of necroptosis and apoptosis by regulating ubiquitination of RIPK1 and mRNA levels of c-FLIP. Proc Natl Acad Sci USA 114: 11944–11949, 2017. doi: 10.1073/pnas.1715742114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 504.Wang H, Sun L, Su L, Rizo J, Liu L, Wang LF, Wang FS, Wang X. Mixed lineage kinase domain-like protein MLKL causes necrotic membrane disruption upon phosphorylation by RIP3. Mol Cell 54: 133–146, 2014. doi: 10.1016/j.molcel.2014.03.003. [DOI] [PubMed] [Google Scholar]
- 505.Wang H, Yu SW, Koh DW, Lew J, Coombs C, Bowers W, Federoff HJ, Poirier GG, Dawson TM, Dawson VL. Apoptosis-inducing factor substitutes for caspase executioners in NMDA-triggered excitotoxic neuronal death. J Neurosci 24: 10963–10973, 2004. doi: 10.1523/JNEUROSCI.3461-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 506.Wang HG, Pathan N, Ethell IM, Krajewski S, Yamaguchi Y, Shibasaki F, McKeon F, Bobo T, Franke TF, Reed JC. Ca2+-induced apoptosis through calcineurin dephosphorylation of BAD. Science 284: 339–343, 1999. doi: 10.1126/science.284.5412.339. [DOI] [PubMed] [Google Scholar]
- 507.Wang JX, Zhang XJ, Li Q, Wang K, Wang Y, Jiao JQ, Feng C, Teng S, Zhou LY, Gong Y, Zhou ZX, Liu J, Wang JL, Li PF. MicroRNA-103/107 Regulate Programmed Necrosis and Myocardial Ischemia/Reperfusion Injury Through Targeting FADD. Circ Res 117: 352–363, 2015. doi: 10.1161/CIRCRESAHA.117.305781. [DOI] [PubMed] [Google Scholar]
- 508.Wang K, Liu F, Liu CY, An T, Zhang J, Zhou LY, Wang M, Dong YH, Li N, Gao JN, Zhao YF, Li PF. The long noncoding RNA NRF regulates programmed necrosis and myocardial injury during ischemia and reperfusion by targeting miR-873. Cell Death Differ 23: 1394–1405, 2016. doi: 10.1038/cdd.2016.28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 509.Wang K, Yin XM, Chao DT, Milliman CL, Korsmeyer SJ. BID: a novel BH3 domain-only death agonist. Genes Dev 10: 2859–2869, 1996. doi: 10.1101/gad.10.22.2859. [DOI] [PubMed] [Google Scholar]
- 510.Wang L, Yang JK, Kabaleeswaran V, Rice AJ, Cruz AC, Park AY, Yin Q, Damko E, Jang SB, Raunser S, Robinson CV, Siegel RM, Walz T, Wu H. The Fas-FADD death domain complex structure reveals the basis of DISC assembly and disease mutations. Nat Struct Mol Biol 17: 1324–1329, 2010. doi: 10.1038/nsmb.1920. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 511.Wang Y, An R, Umanah GK, Park H, Nambiar K, Eacker SM, Kim B, Bao L, Harraz MM, Chang C, Chen R, Wang JE, Kam TI, Jeong JS, Xie Z, Neifert S, Qian J, Andrabi SA, Blackshaw S, Zhu H, Song H, Ming GL, Dawson VL, Dawson TM. A nuclease that mediates cell death induced by DNA damage and poly(ADP-ribose) polymerase-1. Science 354: aad6872, 2016. doi: 10.1126/science.aad6872. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 512.Wang Y, Gao W, Shi X, Ding J, Liu W, He H, Wang K, Shao F. Chemotherapy drugs induce pyroptosis through caspase-3 cleavage of a gasdermin. Nature 547: 99–103, 2017. doi: 10.1038/nature22393. [DOI] [PubMed] [Google Scholar]
- 513.Wang Y, Kim NS, Haince JF, Kang HC, David KK, Andrabi SA, Poirier GG, Dawson VL, Dawson TM. Poly(ADP-ribose) (PAR) binding to apoptosis-inducing factor is critical for PAR polymerase-1-dependent cell death (parthanatos). Sci Signal 4: ra20, 2011. doi: 10.1126/scisignal.2000902. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 514.Ware CF, Crowe PD, Vanarsdale TL, Andrews JL, Grayson MH, Jerzy R, Smith CA, Goodwin RG. Tumor necrosis factor (TNF) receptor expression in T lymphocytes. Differential regulation of the type I TNF receptor during activation of resting and effector T cells. J Immunol 147: 4229–4238, 1991. [PubMed] [Google Scholar]
- 515.Waterhouse NJ, Sedelies KA, Browne KA, Wowk ME, Newbold A, Sutton VR, Clarke CJ, Oliaro J, Lindemann RK, Bird PI, Johnstone RW, Trapani JA. A central role for Bid in granzyme B-induced apoptosis. J Biol Chem 280: 4476–4482, 2005. doi: 10.1074/jbc.M410985200. [DOI] [PubMed] [Google Scholar]
- 516.Waterhouse P, Penninger JM, Timms E, Wakeham A, Shahinian A, Lee KP, Thompson CB, Griesser H, Mak TW. Lymphoproliferative disorders with early lethality in mice deficient in Ctla-4. Science 270: 985–988, 1995. doi: 10.1126/science.270.5238.985. [DOI] [PubMed] [Google Scholar]
- 517.Wayman N, McDonald MC, Thompson AS, Threadgill MD, Thiemermann C. 5-Aminoisoquinolinone, a potent inhibitor of poly (adenosine 5′-diphosphate ribose) polymerase, reduces myocardial infarct size. Eur J Pharmacol 430: 93–100, 2001. doi: 10.1016/S0014-2999(01)01359-0. [DOI] [PubMed] [Google Scholar]
- 518.Weber JS, Kähler KC, Hauschild A. Management of immune-related adverse events and kinetics of response with ipilimumab. J Clin Oncol 30: 2691–2697, 2012. doi: 10.1200/JCO.2012.41.6750. [DOI] [PubMed] [Google Scholar]
- 519.Weckbach LT, Grabmaier U, Uhl A, Gess S, Boehm F, Zehrer A, Pick R, Salvermoser M, Czermak T, Pircher J, Sorrelle N, Migliorini M, Strickland DK, Klingel K, Brinkmann V, Abu Abed U, Eriksson U, Massberg S, Brunner S, Walzog B. Midkine drives cardiac inflammation by promoting neutrophil trafficking and NETosis in myocarditis. J Exp Med 216: 350–368, 2019. doi: 10.1084/jem.20181102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 520.Wei MC, Zong WX, Cheng EH, Lindsten T, Panoutsakopoulou V, Ross AJ, Roth KA, MacGregor GR, Thompson CB, Korsmeyer SJ. Proapoptotic BAX and BAK: a requisite gateway to mitochondrial dysfunction and death. Science 292: 727–730, 2001. doi: 10.1126/science.1059108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 521.Wei R, Xu LW, Liu J, Li Y, Zhang P, Shan B, Lu X, Qian L, Wu Z, Dong K, Zhu H, Pan L, Yuan J, Pan H. SPATA2 regulates the activation of RIPK1 by modulating linear ubiquitination. Genes Dev 31: 1162–1176, 2017. doi: 10.1101/gad.299776.117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 522.Wei SC, Duffy CR, Allison JP. Fundamental Mechanisms of Immune Checkpoint Blockade Therapy. Cancer Discov 8: 1069–1086, 2018. doi: 10.1158/2159-8290.CD-18-0367. [DOI] [PubMed] [Google Scholar]
- 523.Weinlich R, Oberst A, Beere HM, Green DR. Necroptosis in development, inflammation and disease. Nat Rev Mol Cell Biol 18: 127–136, 2017. doi: 10.1038/nrm.2016.149. [DOI] [PubMed] [Google Scholar]
- 524.Wencker D, Chandra M, Nguyen K, Miao W, Garantziotis S, Factor SM, Shirani J, Armstrong RC, Kitsis RN. A mechanistic role for cardiac myocyte apoptosis in heart failure. J Clin Invest 111: 1497–1504, 2003. doi: 10.1172/JCI17664. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 525.Wertz IE, Newton K, Seshasayee D, Kusam S, Lam C, Zhang J, Popovych N, Helgason E, Schoeffler A, Jeet S, Ramamoorthi N, Kategaya L, Newman RJ, Horikawa K, Dugger D, Sandoval W, Mukund S, Zindal A, Martin F, Quan C, Tom J, Fairbrother WJ, Townsend M, Warming S, DeVoss J, Liu J, Dueber E, Caplazi P, Lee WP, Goodnow CC, Balazs M, Yu K, Kolumam G, Dixit VM. Phosphorylation and linear ubiquitin direct A20 inhibition of inflammation. [Erratum in Nature 532: 402, 2016.] Nature 528: 370–375, 2015. doi: 10.1038/nature16165. [DOI] [PubMed] [Google Scholar]
- 526.Wertz IE, O’Rourke KM, Zhou H, Eby M, Aravind L, Seshagiri S, Wu P, Wiesmann C, Baker R, Boone DL, Ma A, Koonin EV, Dixit VM. De-ubiquitination and ubiquitin ligase domains of A20 downregulate NF-kappaB signalling. Nature 430: 694–699, 2004. doi: 10.1038/nature02794. [DOI] [PubMed] [Google Scholar]
- 527.Whelan RS, Kaplinskiy V, Kitsis RN. Cell death in the pathogenesis of heart disease: mechanisms and significance. Annu Rev Physiol 72: 19–44, 2010. doi: 10.1146/annurev.physiol.010908.163111. [DOI] [PubMed] [Google Scholar]
- 528.Whelan RS, Konstantinidis K, Wei AC, Chen Y, Reyna DE, Jha S, Yang Y, Calvert JW, Lindsten T, Thompson CB, Crow MT, Gavathiotis E, Dorn GW II, O’Rourke B, Kitsis RN. Bax regulates primary necrosis through mitochondrial dynamics. Proc Natl Acad Sci USA 109: 6566–6571, 2012. doi: 10.1073/pnas.1201608109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 529.Willeford A, Suetomi T, Nickle A, Hoffman HM, Miyamoto S, Heller Brown J. CaMKIIδ-mediated inflammatory gene expression and inflammasome activation in cardiomyocytes initiate inflammation and induce fibrosis. JCI Insight 3: e97054, 2018. doi: 10.1172/jci.insight.97054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 530.Wu J, Huang Z, Ren J, Zhang Z, He P, Li Y, Ma J, Chen W, Zhang Y, Zhou X, Yang Z, Wu SQ, Chen L, Han J. Mlkl knockout mice demonstrate the indispensable role of Mlkl in necroptosis. Cell Res 23: 994–1006, 2013. doi: 10.1038/cr.2013.91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 531.Xiang SY, Ouyang K, Yung BS, Miyamoto S, Smrcka AV, Chen J, Heller Brown J. PLCε, PKD1, and SSH1L transduce RhoA signaling to protect mitochondria from oxidative stress in the heart. Sci Signal 6: ra108, 2013. doi: 10.1126/scisignal.2004405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 532.Xu D, Jin T, Zhu H, Chen H, Ofengeim D, Zou C, Mifflin L, Pan L, Amin P, Li W, Shan B, Naito MG, Meng H, Li Y, Pan H, Aron L, Adiconis X, Levin JZ, Yankner BA, Yuan J. TBK1 Suppresses RIPK1-Driven Apoptosis and Inflammation During Development and in Aging. Cell 174: 1477–1491.e19, 2018. doi: 10.1016/j.cell.2018.07.041. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 533.Xu K, Tavernarakis N, Driscoll M. Necrotic cell death in C. elegans requires the function of calreticulin and regulators of Ca2+ release from the endoplasmic reticulum. Neuron 31: 957–971, 2001. doi: 10.1016/S0896-6273(01)00432-9. [DOI] [PubMed] [Google Scholar]
- 534.Xu Z, Zhang J, David KK, Yang ZJ, Li X, Dawson TM, Dawson VL, Koehler RC. Endonuclease G does not play an obligatory role in poly(ADP-ribose) polymerase-dependent cell death after transient focal cerebral ischemia. Am J Physiol Regul Integr Comp Physiol 299: R215–R221, 2010. doi: 10.1152/ajpregu.00747.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 535.Yagoda N, von Rechenberg M, Zaganjor E, Bauer AJ, Yang WS, Fridman DJ, Wolpaw AJ, Smukste I, Peltier JM, Boniface JJ, Smith R, Lessnick SL, Sahasrabudhe S, Stockwell BR. RAS-RAF-MEK-dependent oxidative cell death involving voltage-dependent anion channels. Nature 447: 865–868, 2007. doi: 10.1038/nature05859. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 536.Yan Y, Jiang W, Liu L, Wang X, Ding C, Tian Z, Zhou R. Dopamine controls systemic inflammation through inhibition of NLRP3 inflammasome. Cell 160: 62–73, 2015. doi: 10.1016/j.cell.2014.11.047. [DOI] [PubMed] [Google Scholar]
- 537.Yang QH, Church-Hajduk R, Ren J, Newton ML, Du C. Omi/HtrA2 catalytic cleavage of inhibitor of apoptosis (IAP) irreversibly inactivates IAPs and facilitates caspase activity in apoptosis. Genes Dev 17: 1487–1496, 2003. doi: 10.1101/gad.1097903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 538.Yang W, Guastella J, Huang JC, Wang Y, Zhang L, Xue D, Tran M, Woodward R, Kasibhatla S, Tseng B, Drewe J, Cai SX. MX1013, a dipeptide caspase inhibitor with potent in vivo antiapoptotic activity. Br J Pharmacol 140: 402–412, 2003. doi: 10.1038/sj.bjp.0705450. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 539.Yang WS, Kim KJ, Gaschler MM, Patel M, Shchepinov MS, Stockwell BR. Peroxidation of polyunsaturated fatty acids by lipoxygenases drives ferroptosis. Proc Natl Acad Sci USA 113: E4966–E4975, 2016. doi: 10.1073/pnas.1603244113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 540.Yang WS, SriRamaratnam R, Welsch ME, Shimada K, Skouta R, Viswanathan VS, Cheah JH, Clemons PA, Shamji AF, Clish CB, Brown LM, Girotti AW, Cornish VW, Schreiber SL, Stockwell BR. Regulation of ferroptotic cancer cell death by GPX4. Cell 156: 317–331, 2014. doi: 10.1016/j.cell.2013.12.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 542.Yang Y, Fang S, Jensen JP, Weissman AM, Ashwell JD. Ubiquitin protein ligase activity of IAPs and their degradation in proteasomes in response to apoptotic stimuli. Science 288: 874–877, 2000. doi: 10.1126/science.288.5467.874. [DOI] [PubMed] [Google Scholar]
- 543.Yang Z, Zingarelli B, Szabó C. Effect of genetic disruption of poly (ADP-ribose) synthetase on delayed production of inflammatory mediators and delayed necrosis during myocardial ischemia-reperfusion injury. Shock 13: 60–66, 2000. doi: 10.1097/00024382-200013010-00011. [DOI] [PubMed] [Google Scholar]
- 544.Yaoita H, Ogawa K, Maehara K, Maruyama Y. Attenuation of ischemia/reperfusion injury in rats by a caspase inhibitor. Circulation 97: 276–281, 1998. doi: 10.1161/01.CIR.97.3.276. [DOI] [PubMed] [Google Scholar]
- 546.Yeh RW, Sidney S, Chandra M, Sorel M, Selby JV, Go AS. Population trends in the incidence and outcomes of acute myocardial infarction. N Engl J Med 362: 2155–2165, 2010. doi: 10.1056/NEJMoa0908610. [DOI] [PubMed] [Google Scholar]
- 547.Yeh WC, Itie A, Elia AJ, Ng M, Shu HB, Wakeham A, Mirtsos C, Suzuki N, Bonnard M, Goeddel DV, Mak TW. Requirement for Casper (c-FLIP) in regulation of death receptor-induced apoptosis and embryonic development. Immunity 12: 633–642, 2000. doi: 10.1016/S1074-7613(00)80214-9. [DOI] [PubMed] [Google Scholar]
- 548.Yeh WC, de la Pompa JL, McCurrach ME, Shu HB, Elia AJ, Shahinian A, Ng M, Wakeham A, Khoo W, Mitchell K, El-Deiry WS, Lowe SW, Goeddel DV, Mak TW. FADD: essential for embryo development and signaling from some, but not all, inducers of apoptosis. Science 279: 1954–1958, 1998. doi: 10.1126/science.279.5358.1954. [DOI] [PubMed] [Google Scholar]
- 549.Yellon DM, Hausenloy DJ. Myocardial reperfusion injury. N Engl J Med 357: 1121–1135, 2007. doi: 10.1056/NEJMra071667. [DOI] [PubMed] [Google Scholar]
- 550.Yokosuka T, Takamatsu M, Kobayashi-Imanishi W, Hashimoto-Tane A, Azuma M, Saito T. Programmed cell death 1 forms negative costimulatory microclusters that directly inhibit T cell receptor signaling by recruiting phosphatase SHP2. J Exp Med 209: 1201–1217, 2012. doi: 10.1084/jem.20112741. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 551.Yoon S, Kovalenko A, Bogdanov K, Wallach D. MLKL, the Protein that Mediates Necroptosis, Also Regulates Endosomal Trafficking and Extracellular Vesicle Generation. Immunity 47: 51–65.e7, 2017. doi: 10.1016/j.immuni.2017.06.001. [DOI] [PubMed] [Google Scholar]
- 552.Yu SW, Andrabi SA, Wang H, Kim NS, Poirier GG, Dawson TM, Dawson VL. Apoptosis-inducing factor mediates poly(ADP-ribose) (PAR) polymer-induced cell death. Proc Natl Acad Sci USA 103: 18314–18319, 2006. doi: 10.1073/pnas.0606528103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 553.Yu SW, Wang H, Poitras MF, Coombs C, Bowers WJ, Federoff HJ, Poirier GG, Dawson TM, Dawson VL. Mediation of poly(ADP-ribose) polymerase-1-dependent cell death by apoptosis-inducing factor. Science 297: 259–263, 2002. doi: 10.1126/science.1072221. [DOI] [PubMed] [Google Scholar]
- 554.Yuan J, Shaham S, Ledoux S, Ellis HM, Horvitz HR. The C. elegans cell death gene ced-3 encodes a protein similar to mammalian interleukin-1 beta-converting enzyme. Cell 75: 641–652, 1993. doi: 10.1016/0092-8674(93)90485-9. [DOI] [PubMed] [Google Scholar]
- 555.Yuan JY, Horvitz HR. The Caenorhabditis elegans genes ced-3 and ced-4 act cell autonomously to cause programmed cell death. Dev Biol 138: 33–41, 1990. doi: 10.1016/0012-1606(90)90174-H. [DOI] [PubMed] [Google Scholar]
- 556.Yue Z, Jin S, Yang C, Levine AJ, Heintz N. Beclin 1, an autophagy gene essential for early embryonic development, is a haploinsufficient tumor suppressor. Proc Natl Acad Sci USA 100: 15077–15082, 2003. doi: 10.1073/pnas.2436255100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 557.Yussman MG, Toyokawa T, Odley A, Lynch RA, Wu G, Colbert MC, Aronow BJ, Lorenz JN, Dorn GW II. Mitochondrial death protein Nix is induced in cardiac hypertrophy and triggers apoptotic cardiomyopathy. Nat Med 8: 725–730, 2002. doi: 10.1038/nm719. [DOI] [PubMed] [Google Scholar]
- 558.Zargarian S, Shlomovitz I, Erlich Z, Hourizadeh A, Ofir-Birin Y, Croker BA, Regev-Rudzki N, Edry-Botzer L, Gerlic M. Phosphatidylserine externalization, “necroptotic bodies” release, and phagocytosis during necroptosis. PLoS Biol 15: e2002711, 2017. doi: 10.1371/journal.pbio.2002711. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 559.Zhang H, Zhou X, McQuade T, Li J, Chan FK, Zhang J. Functional complementation between FADD and RIP1 in embryos and lymphocytes. Nature 471: 373–376, 2011. doi: 10.1038/nature09878. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 560.Zhang J, Dawson VL, Dawson TM, Snyder SH. Nitric oxide activation of poly(ADP-ribose) synthetase in neurotoxicity. [Corrigendum in Science 483: 498, 2011.] Science 263: 687–689, 1994. doi: 10.1126/science.8080500. [DOI] [PubMed] [Google Scholar]
- 561.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 562.Zhang L, Blackwell K, Shi Z, Habelhah H. The RING domain of TRAF2 plays an essential role in the inhibition of TNFalpha-induced cell death but not in the activation of NF-kappaB. J Mol Biol 396: 528–539, 2010. doi: 10.1016/j.jmb.2010.01.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 563.Zhang T, Zhang Y, Cui M, Jin L, Wang Y, Lv F, Liu Y, Zheng W, Shang H, Zhang J, Zhang M, Wu H, Guo J, Zhang X, Hu X, Cao CM, Xiao RP. CaMKII is a RIP3 substrate mediating ischemia- and oxidative stress-induced myocardial necroptosis. Nat Med 22: 175–182, 2016. doi: 10.1038/nm.4017. [DOI] [PubMed] [Google Scholar]
- 564.Zhao J, Jitkaew S, Cai Z, Choksi S, Li Q, Luo J, Liu ZG. Mixed lineage kinase domain-like is a key receptor interacting protein 3 downstream component of TNF-induced necrosis. Proc Natl Acad Sci USA 109: 5322–5327, 2012. doi: 10.1073/pnas.1200012109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 565.Zhong Z, Liang S, Sanchez-Lopez E, He F, Shalapour S, Lin XJ, Wong J, Ding S, Seki E, Schnabl B, Hevener AL, Greenberg HB, Kisseleva T, Karin M. New mitochondrial DNA synthesis enables NLRP3 inflammasome activation. Nature 560: 198–203, 2018. doi: 10.1038/s41586-018-0372-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 566.Zhong Z, Umemura A, Sanchez-Lopez E, Liang S, Shalapour S, Wong J, He F, Boassa D, Perkins G, Ali SR, McGeough MD, Ellisman MH, Seki E, Gustafsson AB, Hoffman HM, Diaz-Meco MT, Moscat J, Karin M. NF-κB Restricts Inflammasome Activation via Elimination of Damaged Mitochondria. Cell 164: 896–910, 2016. doi: 10.1016/j.cell.2015.12.057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 567.Zhou HZ, Swanson RA, Simonis U, Ma X, Cecchini G, Gray MO. Poly(ADP-ribose) polymerase-1 hyperactivation and impairment of mitochondrial respiratory chain complex I function in reperfused mouse hearts. Am J Physiol Heart Circ Physiol 291: H714–H723, 2006. doi: 10.1152/ajpheart.00823.2005. [DOI] [PubMed] [Google Scholar]
- 568.Zhou R, Yazdi AS, Menu P, Tschopp J. A role for mitochondria in NLRP3 inflammasome activation. [Corrigendum in Nature 475: 122, 2011.] Nature 469: 221–225, 2011. doi: 10.1038/nature09663. [DOI] [PubMed] [Google Scholar]
- 569.Zhou W, Marinelli F, Nief C, Faraldo-Gómez JD. Atomistic simulations indicate the c-subunit ring of the F1Fo ATP synthase is not the mitochondrial permeability transition pore. eLife 6: e23781, 2017. doi: 10.7554/eLife.23781. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 570.Zhu H, Tannous P, Johnstone JL, Kong Y, Shelton JM, Richardson JA, Le V, Levine B, Rothermel BA, Hill JA. Cardiac autophagy is a maladaptive response to hemodynamic stress. J Clin Invest 117: 1782–1793, 2007. doi: 10.1172/JCI27523. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 571.Zhu W, Woo AY, Yang D, Cheng H, Crow MT, Xiao RP. Activation of CaMKIIdeltaC is a common intermediate of diverse death stimuli-induced heart muscle cell apoptosis. J Biol Chem 282: 10833–10839, 2007. doi: 10.1074/jbc.M611507200. [DOI] [PubMed] [Google Scholar]
- 572.Zingarelli B, Cuzzocrea S, Zsengellér Z, Salzman AL, Szabó C. Protection against myocardial ischemia and reperfusion injury by 3-aminobenzamide, an inhibitor of poly (ADP-ribose) synthetase. Cardiovasc Res 36: 205–215, 1997. doi: 10.1016/S0008-6363(97)00137-5. [DOI] [PubMed] [Google Scholar]
- 573.Zingarelli B, Hake PW, O’Connor M, Denenberg A, Kong S, Aronow BJ. Absence of poly(ADP-ribose)polymerase-1 alters nuclear factor-kappa B activation and gene expression of apoptosis regulators after reperfusion injury. Mol Med 9: 143–153, 2003. doi: 10.1007/BF03402179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 574.Zong WX, Li C, Hatzivassiliou G, Lindsten T, Yu QC, Yuan J, Thompson CB. Bax and Bak can localize to the endoplasmic reticulum to initiate apoptosis. J Cell Biol 162: 59–69, 2003. doi: 10.1083/jcb.200302084. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 575.Zou H, Henzel WJ, Liu X, Lutschg A, Wang X. Apaf-1, a human protein homologous to C. elegans CED-4, participates in cytochrome c-dependent activation of caspase-3. Cell 90: 405–413, 1997. doi: 10.1016/S0092-8674(00)80501-2. [DOI] [PubMed] [Google Scholar]