Fact-checked by Grok 2 weeks ago

Cell death

Cell death is an irreversible by which cells lose their ability to maintain vital functions, leading to their demise and subsequent clearance from tissues, and it encompasses both accidental cell death (ACD), triggered by severe injury, and regulated cell death (RCD), a controlled mechanism mediated by dedicated molecular machinery. RCD is essential for embryonic development, adult tissue , and immune responses, as it eliminates superfluous, damaged, or infected cells while minimizing in many cases. The major forms of RCD include , a caspase-dependent process featuring cell shrinkage, condensation, and apoptotic body formation without significant ; necroptosis, an inflammatory lytic death involving receptor-interacting protein kinase 3 (RIPK3) and mixed lineage kinase domain-like protein (MLKL); , triggered by and gasdermin pores leading to release and immune activation; and , an iron-dependent form driven by . These pathways are interconnected and can switch based on cellular context, with dysregulation implicated in diseases such as cancer, neurodegeneration, and infectious disorders.

Overview and Fundamentals

Definition and Characteristics

Cell death is defined as the irreversible degeneration of vital cellular functions, such as ATP production and homeostasis, ultimately leading to the loss of cellular integrity through permanent plasma permeabilization or fragmentation. This process encompasses both regulated cell death (RCD), which involves genetically encoded molecular mechanisms to eliminate superfluous, damaged, or harmful cells in response to physiological or pathological cues, and accidental cell death (ACD), an uncontrolled response to severe physical, chemical, or mechanical insults that causes instantaneous cellular demise. These forms contribute to organismal adaptation during development and or to in diseases like cancer and neurodegeneration. Unlike reversible cellular states, cell death marks a where cannot be restored, distinguishing it from quiescence—a temporary, reversible proliferative in response to nutrient limitation or absence—and , an irreversible withdrawal accompanied by sustained metabolic activity and secretion of pro-inflammatory factors, but without loss of membrane integrity or vital functions. In quiescence, cells retain the potential to re-enter the upon favorable conditions, whereas senescent cells remain viable yet non-proliferative, often serving regulatory roles in tissue repair or tumor suppression. General morphological characteristics of cell death include cell body shrinkage, plasma membrane blebbing, organelle swelling or dysfunction, and nuclear alterations such as chromatin condensation and fragmentation, which collectively signal the breakdown of structural integrity across various forms. Biochemically, hallmarks encompass the depletion of cellular ATP, activation of degradative proteases, and progressive loss of membrane asymmetry and integrity, often detected by assays measuring dysregulation or extracellular release of intracellular contents. Cell death processes exhibit evolutionary conservation across eukaryotes, from unicellular organisms like and Dictyostelium discoideum to multicellular animals and , involving shared elements such as mitochondrial involvement and apoptosis-inducing factor (AIF) homologs that facilitate programmed elimination for population-level . This antiquity suggests that core mechanisms arose early in eukaryotic evolution, predating complex multicellularity, and were co-opted for developmental and stress-response roles.

Historical Development

The concept of cell death at the cellular level emerged in the mid-19th century through pathological observations. In 1858, , in his seminal work Die Cellularpathologie, described as a form of tissue death attributable to individual cell alterations during and injury, marking the first recognition of cell death as a distinct pathological process rather than a mere consequence of organismal decay. This laid the groundwork for understanding cell death as a cellular phenomenon, shifting focus from to microscopic changes. The 20th century brought systematic studies distinguishing regulated from accidental cell death. In 1972, John F.R. Kerr, Andrew H. Wyllie, and Alastair R. Currie coined the term "" to describe a morphologically distinct, non-inflammatory form of cell death observed in normal and pathological tissues, differentiating it from the disruptive . Concurrently, research on revealed as an active process; in the 1980s, John E. Sulston and mapped the cell lineage, identifying 113 cells that undergo genetically controlled death during embryogenesis (contributing to the total of 131 deaths in development), establishing as essential for normal development. Advances in the 1980s and 1990s uncovered key molecular regulators. The protein was identified in 1988 as an product from the t(14;18) translocation in follicular lymphomas, later recognized in 1988-1990 studies as an , revealing anti-death mechanisms in cancer. , a family of cysteine proteases, were first linked to cell death in 1993 when Junying Yuan and colleagues showed that the C. elegans gene ced-3 encodes a protease homologous to mammalian interleukin-1β-converting enzyme (ICE), central to apoptotic execution. By the early 2000s, regulated necrosis was identified; in 2005, Alexei Degterev and colleagues described necroptosis as a - and RIPK3-dependent pathway, expanding cell death beyond to include inflammatory forms. Recent decades have unveiled additional regulated death modalities, reflecting ongoing paradigm shifts from viewing necrosis solely as passive injury to multifaceted regulated processes. , an iron-dependent lipid peroxidation-driven death, was defined in 2012 by Scott J. Dixon, Brent R. Stockwell, and colleagues as distinct from and . Cuproptosis, triggered by copper overload and tricarboxylic acid cycle disruption, was reported in 2022 by Peter Tsvetkov et al. PANoptosis, integrating , , and necroptosis into an inflammasome-driven response, emerged from 2019 studies by Thirumala-Devi Kanneganti's group. In 2023, disulfidptosis was identified as a novel form of cell death induced by disulfide stress leading to collapse. These discoveries culminated in the 2002 Nobel Prize in Physiology or Medicine awarded to , , and John E. Sulston for elucidating genetic regulation of organ development and via C. elegans research.

Molecular Mechanisms

Initiation Pathways

Cell death initiation pathways encompass the upstream signaling events that detect cellular or external cues and the commitment to programmed cell demise. For , a key form of regulated cell death (RCD), this occurs primarily through two major routes: the extrinsic and intrinsic pathways. These pathways integrate diverse stressors to activate effector mechanisms, ensuring that damaged or unnecessary cells are eliminated in a controlled manner. Other RCD forms, such as necroptosis, , and , involve specialized initiation pathways, including RIPK1/RIPK3 activation for necroptosis, assembly for , and inhibition of glutathione peroxidase 4 (GPX4) leading to for (detailed in the Types of Cell Death section). The extrinsic pathway is activated by extracellular signals via death receptors, while the intrinsic pathway responds to internal perturbations, often converging on mitochondrial dysfunction. Cross-talk between these routes amplifies the signal, highlighting the interconnected of cell death initiation. The extrinsic pathway begins with the ligation of death receptors on the cell surface, such as (TNFR1), (CD95), or TNF-related apoptosis-inducing ligand (TRAIL) receptors, by their respective s. This binding recruits adaptor proteins like (FADD) and procaspase-8 to form the (DISC), leading to the autocatalytic activation of as an initiator . activation propagates the death signal, either directly or by engaging the intrinsic pathway, and this process is tightly regulated to prevent unintended cell loss. In immune , this pathway enables cytotoxic lymphocytes to induce death in target cells expressing . In contrast, the intrinsic pathway is triggered by intracellular stresses, culminating in mitochondrial outer membrane permeabilization (MOMP), a critical checkpoint where pro-apoptotic Bcl-2 family members like Bax and Bak form pores in the mitochondrial membrane. This releases cytochrome c into the cytosol, initiating downstream events, and is often provoked by DNA damage, endoplasmic reticulum (ER) stress, or growth factor withdrawal. The tumor suppressor p53 plays a pivotal role in this pathway by sensing DNA damage through ataxia-telangiectasia mutated (ATM) kinase activation, stabilizing p53 to transcriptionally upregulate pro-apoptotic genes such as Puma and Noxa, which promote MOMP. ER stress, arising from unfolded protein accumulation, similarly activates the unfolded protein response, which, if unresolved, signals through pathways like IRE1 or PERK to induce p53-dependent apoptosis. Common initiators of cell death across pathways include , calcium overload, and deprivation, which disrupt cellular and sensitize cells to death signals. from (ROS) damages DNA, proteins, and lipids, activating and regulators to drive MOMP or receptor sensitization. Calcium overload, often from ER-mitochondria calcium transfer, opens the , exacerbating ROS production and promoting permeabilization independent of Bax/Bak in some contexts. deprivation, such as glucose or scarcity, impairs ATP production and activates (AMPK), which can intersect with to initiate death when survival adaptations fail. Cross-talk between extrinsic and intrinsic pathways is mediated by caspase-8 cleavage of Bid, a BH3-only protein, generating truncated Bid (tBid) that translocates to mitochondria to activate Bax and Bak, thereby linking death receptor signaling to MOMP. This amplification ensures robust commitment to death in cells with weak intrinsic priming. Such integration allows extrinsic signals to harness mitochondrial amplification, enhancing efficiency in contexts like immune-mediated clearance.

Execution and Regulation

The execution of cell death involves intricate proteolytic cascades that amplify initial signals into irreversible commitment. In apoptotic pathways, initiator caspases, such as or , are activated by upstream complexes and subsequently cleave and activate effector caspases like caspase-3 and caspase-7, forming a cascading amplification that dismantles cellular structures through targeted of substrates including cytoskeletal proteins and enzymes. In non-apoptotic forms, such as necroptosis or lysoptosis, execution can proceed via different mechanisms; for necroptosis, receptor-interacting protein 3 (RIPK3) phosphorylates mixed kinase domain-like (MLKL), inducing a conformational change that oligomerizes MLKL and disrupts membrane integrity, serving as a switch when apoptotic are inhibited. For lysoptosis, lysosomal membrane permeabilization (LMP) leads to rupture and release of cathepsins into the , which trigger further proteolytic damage and independent of . In , gasdermin pores form in the membrane following activation, releasing cytokines and causing cell lysis. execution involves accumulation of lipid hydroperoxides leading to membrane rupture. These mechanisms ensure precise control, where the balance of activation thresholds determines whether damage leads to controlled dismantling or lytic disruption. Regulatory proteins finely tune this process by counteracting pro-death signals. Inhibitors of apoptosis proteins (IAPs), particularly XIAP, bind directly to activated -3, -7, and -9, inhibiting their activity through ligase-mediated degradation or steric hindrance, thereby preventing premature execution. Similarly, the integrates pro- and anti-apoptotic signals at the mitochondria; anti-apoptotic members like sequester BH3-only activators, while pro-apoptotic Bax and Bak oligomerize to permeabilize the outer mitochondrial membrane, releasing to activate —yet this is modulated by the relative abundance of family members to maintain cellular . Such regulation allows cells to survive transient stresses, like TNF signaling, by tipping the balance toward survival until damage accumulates beyond repair. Regulators for other RCD forms include ubiquitination for necroptosis suppression and /selenoproteins for inhibition. Feedback loops further integrate execution with adaptive responses, creating dynamic switches between death modalities. For instance, can inhibit through the interaction of Beclin-1 with , where binding sequesters Beclin-1, suppressing formation and thereby reducing cellular clearance that might otherwise promote survival; dissociation of this complex under stress shifts toward pro-death . These loops ensure or cooperation among pathways, preventing aberrant activation. Transcriptional and epigenetic mechanisms, exemplified by , provide long-term regulation favoring survival. Upon activation, translocates to the and induces expression of anti-apoptotic genes such as and IAPs, while epigenetic modifications like histone acetylation enhance its accessibility to promoters, reinforcing a pro-survival transcriptional program that counters death signals. Epigenetic silencing of targets can lower the death threshold, but sustained activity epigenetically maintains cellular resilience against stressors. Cell death commitment often follows threshold models where accumulated damage—such as or protein misfolding—must exceed a critical level to overwhelm regulatory buffers like or IAPs, leading to irreversible execution; below this threshold, repair mechanisms predominate, allowing survival and adaptation. These models highlight how quantitative imbalances in pro- versus anti-death factors dictate , integrating damage with deterministic signaling for robust decision-making.

Types of Cell Death

Apoptosis

Apoptosis represents the canonical form of , characterized by an orderly dismantling of cellular components that ensures non-inflammatory clearance by surrounding cells. This process is essential for maintaining tissue and eliminating superfluous or damaged cells without disrupting neighboring structures. Unlike accidental cell death, apoptosis is genetically regulated and energy-dependent, requiring ATP to execute its morphological and biochemical changes. Morphologically, apoptosis begins with cell shrinkage due to cytoskeletal alterations and loss of cytoplasmic volume, accompanied by condensation into dense, crescent-shaped aggregates adjacent to the . The nucleus undergoes , followed by DNA fragmentation into oligonucleosomal units, producing a characteristic "laddering" pattern visible on . Ultimately, the cell partitions into membrane-bound apoptotic bodies containing intact organelles and nuclear fragments, which are rapidly phagocytosed. These changes distinguish from other cell death modalities by preserving cellular integrity until engulfment. Biochemically, the intrinsic pathway predominates in many apoptotic scenarios, initiated by mitochondrial outer membrane permeabilization (MOMP) that releases into the . binds Apaf-1 and procaspase-9 to form the , which autoactivates ; this initiator then cleaves and activates executioner such as caspase-3, leading to of key substrates like PARP and . The extrinsic pathway can also trigger via death receptors, converging on activation. This cascade ensures precise, irreversible commitment to without widespread degradation seen in other processes. A hallmark of apoptosis is the externalization of (PS) on the outer plasma membrane leaflet, mediated by scramblase activation and inhibited activity. This "eat me" signal promotes by macrophages and neighboring cells through recognition by receptors like TIM-4 or stabilin-2, preventing secondary and . In embryonic development, sculpts structures such as digit separation in the developing limb, where interdigital webs regress through targeted cell elimination.

Autophagy

Autophagy is a conserved eukaryotic process primarily serving cytoprotective functions by degrading and damaged cellular components, but under conditions of prolonged , it can execute . In its lethal form, contributes to demise through excessive self-digestion, distinct from other death pathways by its reliance on lysosomal degradation rather than proteolytic dismantling. This dual role positions as a regulator bridging survival and , particularly in nutrient deprivation or cellular scenarios. The core process of macroautophagy, the most studied type, begins with the formation of a double-membrane structure called the phagophore at the or other membrane sources, which expands to engulf cytoplasmic material, organelles, or protein aggregates. This elongating phagophore matures into an , a vesicle that sequesters the cargo, and subsequently fuses with lysosomes to form an autolysosome, where hydrolytic enzymes degrade the contents into reusable building blocks like and . Key molecular players include autophagy-related (ATG) proteins, such as ATG5 and ATG7, which facilitate phagophore and , while microtubule-associated protein 1 light chain 3 (LC3) undergoes lipidation (LC3-I to LC3-II conversion) to anchor to the autophagosomal membrane, enabling cargo recognition and closure. Initiation is often triggered by inhibition of the mechanistic target of rapamycin () complex 1 under nutrient scarcity, which relieves repression on the ULK1/ATG1 complex to activate downstream ATG machinery. Autophagy encompasses three main types, differentiated by their mechanisms of cargo delivery to lysosomes. Macroautophagy involves the de novo formation of autophagosomes for bulk or selective engulfment, as detailed above, and is the predominant form implicated in both survival and death contexts. Microautophagy entails direct or protrusion of lysosomal or endosomal membranes to internalize small portions of , bypassing intermediate vesicles, and is less characterized in mammalian cell death. (CMA) selectively targets proteins bearing a KFERQ-like via heat shock cognate 70 (HSC70) chaperone recognition, translocating them across the lysosomal membrane through LAMP2A receptor interaction, without vesicle formation. In death execution, excessive macroautophagy can lead to autosis, an autophagy-dependent, non-apoptotic form of cell death characterized by Na+/K+-ATPase pump inhibition, resulting in ionic imbalance, cell swelling, and plasma membrane rupture. Autosis arises from hyperactivation of flux, often induced by or high concentrations of autophagy inducers like Tat-Beclin 1 , overwhelming cellular and depleting essential ions. Unlike typical cytoprotective autophagy, which recycles nutrients to promote survival, prolonged autophagic activity in autosis erodes membrane integrity without inflammatory signaling. Primarily cytoprotective, autophagy becomes lethal under sustained stress, such as or , where it shifts from adaptive recycling to destructive over-degradation, contrasting 's irreversible commitment via activation. shares regulators like Beclin-1 with , allowing crosstalk where autophagic inhibition can sensitize cells to apoptotic signals.

Necrosis

Necrosis represents an unregulated form of death characterized by passive cellular disintegration in response to severe . Morphologically, it involves rapid swelling (oncosis) of the body and organelles, such as mitochondria and the , followed by rupture of the plasma membrane and uncontrolled leakage of intracellular contents into the . This contrasts with more orderly death processes, leading to immediate disruption of architecture. Common triggers of necrosis include acute insults like ischemia (oxygen deprivation), exposure to toxins, or physical , which overwhelm cellular . These stressors typically cause a rapid depletion of ATP through impaired mitochondrial function and , coupled with excessive influx of calcium into the . The resulting energy failure prevents active maintenance of ion gradients and integrity, accelerating the pathological . At the biochemical level, necrosis is marked by the opening of the (mPTP), which dissipates the proton motive force and exacerbates ATP loss while allowing the release of pro-death signals. Concurrently, a burst of (ROS) from dysfunctional mitochondria oxidizes lipids, proteins, and DNA, further promoting organelle damage and membrane permeabilization. The rupture in necrosis releases damage-associated molecular patterns (DAMPs), such as and ATP, which act as endogenous danger signals to trigger sterile inflammation. These DAMPs bind to receptors like Toll-like receptors (TLRs) on immune cells, activating signaling and production to amplify the local inflammatory response. Unlike regulated cell death modalities, classical is accidental and energy-independent, driven solely by overwhelming damage rather than genetic programming; however, under certain conditions, it can transition to regulated variants like necroptosis.

Other Regulated Forms

is an iron-dependent form of regulated cell death characterized by the accumulation of lipid peroxides and (ROS), leading to oxidative damage in cellular membranes. First identified in , is triggered by the inhibition of (GPX4), which normally reduces lipid hydroperoxides; without GPX4 activity, iron-catalyzed Fenton reactions propagate , culminating in plasma membrane rupture. Unlike , is non-apoptotic and pro-inflammatory, with implications in cancer therapy where inducing selectively targets tumor cells reliant on high iron metabolism. Pyroptosis represents a lytic, inflammatory primarily occurring in , activated through signaling that detects microbial or damage-associated patterns. The process involves caspase-1 or caspase-11 cleavage of gasdermin D (GSDMD), releasing its N-terminal fragment, which forms pores in the plasma membrane, causing efflux, swelling, and while facilitating interleukin-1β (IL-1β) . This mechanism, elucidated in 2015, underscores pyroptosis's role in host defense against infections but also contributes to excessive inflammation in and . Necroptosis serves as a regulated backup to , executed when is inhibited, involving the receptor-interacting protein kinases and RIPK3, which phosphorylate mixed lineage kinase domain-like protein (MLKL). Upon , MLKL oligomerizes and translocates to the , disrupting its through formation and leading to a necrotic with . Discovered in 2005 and mechanistically detailed in 2012, necroptosis is often triggered by (TNF) signaling in innate immune contexts, highlighting its importance in antiviral responses and ischemia-reperfusion injury. Cuproptosis, a recently identified -dependent cell death modality, arises from excess intracellular binding to lipoylated proteins in the tricarboxylic acid () cycle, such as dihydrolipoamide S-acetyltransferase (DLAT), causing their aggregation and proteotoxic stress. Described in , this process disrupts mitochondrial respiration and the cycle, leading to abnormal protein accumulation and eventual cell demise distinct from or , with potential therapeutic relevance in copper-dysregulated cancers. Disulfidptosis is a disulfide stress-induced form of regulated cell death, distinct from other modalities, characterized by rapid collapse of the due to aberrant disulfide bond formation in actin fibers under glucose starvation conditions in cells with high expression of the cystine transporter SLC7A11. Identified in , disulfidptosis involves depletion of NADPH and accumulation of disulfides, leading to proteotoxic stress and cell death without activation of typical apoptotic or necroptotic pathways. This mechanism has gained attention for its role in cancer , where inhibiting SLC7A11 can induce disulfidptosis in tumor cells dependent on cystine . PANoptosis integrates features of , , and necroptosis into a unified inflammatory cell death pathway, regulated by the PANoptosome complex involving binding protein 1 (ZBP1), RIP kinases, and in innate immune cells. First proposed in 2019, PANoptosis is activated by viral or bacterial infections, where it promotes coordinated lytic and non-lytic death to enhance antimicrobial immunity, though dysregulated PANoptosis exacerbates inflammatory pathologies like COVID-19-associated storms.

Physiological and Pathological Roles

In Normal Development and

Cell death plays a crucial role in normal development and homeostasis by eliminating superfluous cells, shaping tissues, and maintaining physiological balance. During embryonic development, programmed cell death, particularly , sculpts complex structures by removing transient cell populations that are no longer needed. For instance, in limb development, apoptosis in the interdigital mesenchyme leads to the regression of tissue between digits, forming separated fingers and toes. This process is highly conserved across species, as evidenced by the precise elimination of 131 somatic cells out of 1,090 generated during Caenorhabditis elegans hermaphrodite development, which refines the and other structures. In tissue homeostasis, cell death ensures the renewal of cell populations by balancing proliferation and turnover, particularly through the removal of senescent or damaged cells via . A striking example is the maturation of lens fiber cells, where programmed organelle degradation and nuclear elimination create an organelle-free zone essential for lens transparency and visual clarity. Similarly, during erythroid maturation, enucleation expels the nucleus from precursor cells, enabling the production of functional, biconcave erythrocytes without nuclear remnants, a process involving unconventional cell death mechanisms that support oxygen transport efficiency. In the immune system, is vital for thymic selection, where over 95% of developing T lymphocytes undergo death to eliminate self-reactive clones, thereby establishing central tolerance and preventing . Cell death also contributes to immune homeostasis by regulating lymphocyte survival and pathogen defense. Autophagy promotes T-cell survival and differentiation by degrading organelles and maintaining metabolic balance, which is essential for sustaining adaptive immune responses without exhaustion. Meanwhile, regulated necrosis, such as necroptosis, facilitates viral clearance by inducing inflammatory cell death in infected cells, alerting the and limiting spread under physiological conditions. Quantitatively, these processes are immense in scale; approximately 10<sup>11</sup> cells undergo daily in an adult human to support renewal and , equivalent to replacing the entire body's mass over time.

In Disease Processes

Dysregulation of cell death pathways plays a central role in cancer progression, where evasion of apoptosis confers a survival advantage to malignant cells. Overexpression of the anti-apoptotic protein Bcl-2 is a hallmark mechanism of apoptosis resistance in various cancers, including lymphomas and solid tumors, allowing cancer cells to evade chemotherapy-induced death. This resistance arises from Bcl-2's inhibition of pro-apoptotic effectors like Bax and Bak, preserving mitochondrial integrity and preventing cytochrome c release. Emerging evidence also highlights ferroptosis, an iron-dependent form of regulated necrosis, as a vulnerability in cancer cells; therapies targeting ferroptosis, such as inhibitors of glutathione peroxidase 4 (GPX4), exploit lipid peroxidation to selectively kill tumor cells resistant to apoptosis. In neurodegenerative diseases, excessive activation of necroptosis contributes to neuronal loss and pathology. In , receptor-interacting protein kinase 1 () hyperactivation drives necroptosis in , exacerbating amyloid-beta plaque formation and hyperphosphorylation, leading to cognitive decline. Similarly, failure of in impairs the clearance of aggregates, resulting in death and motor deficits; this autophagic dysfunction stems from lysosomal impairments and mitochondrial abnormalities. These dysregulations underscore how uncontrolled cell death amplifies and in these conditions. Cardiovascular diseases involve dysregulated and , promoting tissue damage and inflammation. Uncontrolled during leads to cardiomyocyte death following ischemia-reperfusion injury, expanding infarct size and impairing cardiac function through release of damage-associated molecular patterns (DAMPs). In atherosclerosis, of vascular endothelial cells and macrophages, mediated by the and gasdermin D, amplifies plaque instability and inflammatory responses, accelerating lesion progression and rupture risk. In infectious diseases, cell death pathways serve dual roles in host defense and pathogen persistence. Host cell , triggered by activation during bacterial infections, limits intracellular pathogen spread by lysing infected cells and releasing antimicrobial contents, as seen in responses to and . Conversely, many viruses inhibit to prolong host cell survival and facilitate replication; for instance, herpesviruses and adenoviruses encode homologs that block mitochondrial outer membrane permeabilization, evading immune clearance. Therapeutic strategies targeting dysregulated cell death hold promise across these diseases. BH3 mimetics, such as , restore in cancers with overexpression by competitively binding anti-apoptotic proteins, inducing tumor cell death and showing efficacy in . For ferroptosis-related pathologies, agonists or stabilizers suppress excessive , potentially mitigating neurodegeneration and ischemia-reperfusion injury in cardiovascular events. Additionally, emerging research links cuproptosis—a copper-dependent cell death involving tricarboxylic acid cycle disruption—to metabolic diseases like , where copper dysregulation in mitochondria may contribute to beta-cell loss and , suggesting copper chelators as novel interventions.

Detection and Research Methods

Biochemical and Morphological Assays

Biochemical and morphological assays are essential tools for detecting and characterizing cell death pathways, relying on the identification of structural alterations and enzymatic changes associated with processes like , , and . These methods provide endpoint readouts that distinguish between different forms of cell death based on hallmarks such as DNA fragmentation, membrane permeability, and protein modifications. Morphological assays visualize or cellular changes under , while biochemical assays quantify molecular markers through enzymatic reactions or immunoassays, offering complementary insights into cell death mechanisms. Morphological assays detect structural features of dying cells, such as condensation or cytoplasmic swelling. The Terminal deoxynucleotidyl transferase dUTP nick end labeling () assay identifies DNA double-strand breaks characteristic of by incorporating labeled into fragmented DNA ends, enabling visualization via or in fixed tissues or cells. This method is widely used for detection of apoptotic cells in histological sections, though it can also label DNA breaks from or other insults if not combined with additional markers. Hematoxylin and eosin (H&E) staining, a standard histological technique, reveals through cellular swelling, loss of detail, and eosinophilic in affected tissues, providing a qualitative assessment of necrotic areas in paraffin-embedded samples. Biochemical assays measure the release or activity of intracellular components indicative of membrane compromise or proteolytic cascades. Lactate dehydrogenase (LDH) release assay quantifies cytotoxicity by detecting LDH enzyme leakage from cells with compromised plasma membranes, a hallmark of necrosis or late-stage apoptosis, using colorimetric or luminescent substrates that produce measurable signals proportional to cell death. Caspase activity fluorometry specifically assesses apoptosis by monitoring the proteolytic cleavage of fluorogenic substrates like DEVD-AMC by effector caspases (e.g., caspase-3), generating fluorescent products detectable by spectrofluorometry, with increased activity confirming executioner phase activation in apoptotic pathways. For autophagy, biochemical and morphological markers focus on autophagosome formation and lysosomal activity. blotting for LC3-II, the lipidated form of 1 light chain 3, detects autophagosome accumulation as a shifted band on gels, serving as a for autophagic when combined with lysosomal inhibitors to distinguish synthesis from degradation. LysoTracker dyes, which fluoresce in acidic compartments, stain autolysosomes to morphologically assess autophagy via or , highlighting increased lysosomal acidification during autophagic progression. In necrosis, high mobility group box 1 () release quantification by measures passive extrusion of this nuclear protein from necrotic cells, acting as a that differentiates from non-lytic cell death forms. Flow cytometry using Annexin V conjugated to fluorophores (e.g., FITC) combined with propidium iodide (PI) provides a dual-staining approach to differentiate stages: V binds externalized on early apoptotic cells with intact membranes, while PI enters late apoptotic or cells with permeabilized membranes, allowing quantitative distinction of viable, early apoptotic, late apoptotic, and populations in heterogeneous samples. This method's advantages include high throughput and sensitivity for mixed cell death scenarios, but limitations arise from specificity issues, such as V binding to non-apoptotic cells under certain stresses or PI's inability to detect early without additional markers.

Molecular Imaging and Genetic Tools

Molecular imaging techniques enable real-time visualization of cell death processes in living cells and tissues. Live-cell utilizing (FRET)-based sensors for has revolutionized the study of by allowing dynamic monitoring of activation. These genetically encoded probes consist of a caspase-cleavable linker between a donor (e.g., CFP) and an acceptor (e.g., YFP), where cleavage disrupts energy transfer, increasing the donor-to-acceptor emission ratio upon caspase engagement. For instance, FRET sensors targeting caspase-3 have detected early apoptotic signaling in neuronal models, providing spatiotemporal resolution of executioner caspase activity before morphological changes occur. Similarly, multiplexed FRET bioprobes have facilitated combinatorial imaging of caspase cascades in cell populations, revealing heterogeneous apoptotic responses to stimuli. Super-resolution microscopy has advanced the imaging of necroptosis by resolving nanoscale structures like mixed-lineage kinase domain-like (MLKL) pores in the plasma membrane. has visualized phosphorylated MLKL oligomerization and translocation to and polymerization on lysosomal or plasma membranes during necroptotic execution, demonstrating MLKL's role in membrane disruption and distinguishing it from other lytic deaths. In addition, of necrosomes has shown that RIPK3 oligomers of tetrameric size or larger recruit MLKL, highlighting critical checkpoints in necroptosis initiation. Genetic tools, including /Cas9-mediated knockouts, have been instrumental in dissecting and necroptosis pathways. knockout of autophagy-related (ATG) genes, such as ATG5 or ATG7, blocks autophagosome formation and flux, enabling precise evaluation of 's role in cell death modulation; for example, ATG7 deletion in lines has revealed its essentiality in mitophagy during stress-induced death. Genome-wide screens using ATG-targeted libraries have identified novel regulators of selective , confirming core ATG factors while uncovering organelle-specific modifiers. For necroptosis, conditional RIPK3 floxed mouse models allow tissue-specific deletion, demonstrating RIPK3's protective roles in independent of necroptosis in macrophages without global lethality. These models have shown that endothelial RIPK3 deletion exacerbates plaque progression, underscoring cell-type-specific contributions to inflammatory processes. Omics approaches provide high-throughput insights into cell death dynamics. Single-cell RNA sequencing (scRNA-seq) reconstructs death trajectories by pseudotime analysis, mapping transcriptional changes from initiation to execution; for instance, scRNA-seq in senescent cells has identified regulators of and subroutines via Death-seq, a method enriching dying cells for pathway screening. has systematically identified substrates, with proteome-wide screens revealing over 1,000 cleavage sites across apoptotic contexts, including family-specific motifs for caspases-3 and -7 that exclude certain proteins to fine-tune death signaling. These tools emphasize substrate prioritization, such as PARP1 and lamin A, in executioner phases. Emerging techniques integrate and (AI) for precise control and analysis. Optogenetic systems induce via light-activated oligomerization of pro-apoptotic proteins like Bax or ; blue light triggers mitochondrial recruitment of opto-Bax, initiating release within minutes in mammalian cells. Similarly, light-inducible constructs for RIPK3-MLKL complexes enable necroptosis activation, offering spatiotemporal precision . AI-assisted analysis enhances detection by classifying lipid (ROS) accumulation; models trained on datasets distinguish ferroptotic morphology from with over 90% accuracy, quantifying via on TfR1-stained images. Recent 2020s advancements include for tissue-level cell death profiling. Techniques like Visium or NanoString GeoMx map death-related , revealing microenvironments promoting turnover; for example, profiling shed cells from the tract has quantified epithelial turnover rates and identified pro-inflammatory microenvironments associated with short-lived colonocytes (as of 2025). This approach integrates with scRNA-seq to dissect heterogeneous death landscapes in development and .

References

  1. [1]
    A guide to cell death pathways - Nature
    Dec 18, 2023 · In this Review, we provide an overview of the major cell death mechanisms, highlighting the latest insights into their complex regulation and execution.
  2. [2]
    Different types of cell death and their shift in shaping disease - NIH
    Aug 4, 2023 · The researches focus on apoptosis, pyroptosis, necroptosis and autophagy mainly, which all belong to programmed cell death (PCD).
  3. [3]
    Molecular mechanisms of cell death: recommendations of ... - Nature
    Jan 23, 2018 · We propose an updated classification of cell death subroutines focusing on mechanistic and essential (as opposed to correlative and dispensable) aspects of the ...
  4. [4]
    Mechanisms of Cellular Senescence: Cell Cycle Arrest ... - Frontiers
    Cellular senescence is different from another form of growth arrest known as quiescence, in that senescence occurs in G1 and possibly G2 phase of the cell ...Introduction · Senescence Mediated Cell... · The Senescence Associated...
  5. [5]
    On the Evolutionary Conservation of the Cell Death Pathway
    Our findings indicate that the cell death pathway in Dictyostelium involves mitochondria and an AIF homolog, suggesting the evolutionary conservation of at ...
  6. [6]
  7. [7]
  8. [8]
    Diversity and complexity of cell death: a historical review - Nature
    Aug 23, 2023 · In this review, we provide a historical overview of the discovery and characterization of different forms of cell death and highlight their diversity and ...
  9. [9]
    The concept of intrinsic versus extrinsic apoptosis - Portland Press
    The extrinsic apoptotic pathway in mammals follows two main routes according to different cell types. In the type I death receptor pathway, cells undergo ...
  10. [10]
    Cross-Talk in Cell Death Signaling - PMC - NIH
    ... cross-talk occurs between the extrinsic and intrinsic pathways in vivo. At a molecular level, this appears to occur via the proteolysis of BID, which ...
  11. [11]
    Cell Death: Critical Control Points - ScienceDirect.com
    Jan 23, 2004 · For example, the extrinsic pathway is triggered by the engagement of cell surface death receptors, which then activate caspase-8 that cleaves ...
  12. [12]
    Mitochondrial outer membrane permeabilization during apoptosis
    May 19, 2006 · In both situations, caspase-dependent and -independent cell death, MOMP occurs. This disrupts mitochondrial function and, even in the absence of ...
  13. [13]
    The Cell-Cycle Arrest and Apoptotic Functions of p53 in Tumor ...
    p53 most notably induces cell-cycle arrest and cell death, thereby blocking tumor progression. But p53 also regulates metabolism, antioxidant response, and DNA ...
  14. [14]
    Oxidative cell death in cancer: mechanisms and therapeutic ... - Nature
    Aug 1, 2024 · This review provides a comprehensive summary of recent progress, opportunities, and challenges in targeting oxidative cell death for cancer therapy.Missing: deprivation | Show results with:deprivation
  15. [15]
    Cell death as a result of calcium signaling modulation: A cancer ...
    In this work, we review the latest useful evidence to better understand the complex network of pathways by which Ca 2+ regulates cell life and death decisions.
  16. [16]
    Regulated cell death pathways and their roles in homeostasis ...
    Aug 23, 2023 · The proteolytic cascade leading to activation of caspases is one of the biochemical features of both extrinsic and intrinsic apoptosis.
  17. [17]
    Lysoptosis is an evolutionarily conserved cell death pathway ...
    Jan 12, 2022 · The role of lysosomes and LMP in mammalian cell death pathways has been difficult to ascertain. LMP and cathepsin release are detected in most ...
  18. [18]
    Regulation of apoptosis by XIAP ubiquitin-ligase activity
    Inhibitor of Apoptosis Proteins (IAPs) can bind to and inhibit caspases, the key executioners of apoptosis. Because IAPs are frequently overexpressed in human ...
  19. [19]
    The BCL2 family: from apoptosis mechanisms to new advances in ...
    Mar 21, 2025 · The B cell lymphoma 2 (BCL2) protein family critically controls apoptosis by regulating the release of cytochrome c from mitochondria.
  20. [20]
    Bcl-2 Antiapoptotic Proteins Inhibit Beclin 1-Dependent Autophagy
    Sep 23, 2005 · Thus, Bcl-2 not only functions as an antiapoptotic protein, but also as an antiautophagy protein via its inhibitory interaction with Beclin 1.
  21. [21]
    The NF-κB Family of Transcription Factors and Its Regulation - PMC
    NF-κB consists of a family of transcription factors that play critical roles in inflammation, immunity, cell proliferation, differentiation, and survival.
  22. [22]
    Deciphering Programmed Cell Death Rules for Cancer Therapy
    The Bcl-2 family can be regarded as a tripartite switch that sets the threshold for commitment to cell death, primarily by interactions within the family.
  23. [23]
    Apoptosis: A Basic Biological Phenomenon with Wide-ranging ... - NIH
    Apoptosis: A Basic Biological Phenomenon with Wide-ranging Implications in Tissue Kinetics · J F R Kerr · A H Wyllie · A R Currie.
  24. [24]
    Induction of apoptotic program in cell-free extracts - PubMed
    Cells undergoing apoptosis in vivo showed increased release of cytochrome c to their cytosol, suggesting that mitochondria may function in apoptosis by ...Missing: seminal paper
  25. [25]
    Autophagy: Regulator of cell death | Cell Death & Disease - Nature
    Oct 4, 2023 · Autophagy is the process by which cells degrade and recycle proteins and organelles to maintain intracellular homeostasis.
  26. [26]
    Autosis is a Na+,K+-ATPase–regulated form of cell death ... - PNAS
    We show that the selective overactivation of autophagy can cause cell death with unique morphological features distinct from apoptosis or necrosis.
  27. [27]
    Autophagy functions in programmed cell death - PMC - NIH
    We show that under physiological conditions during development, autophagy and the Atg genes are required for programmed cell death.
  28. [28]
    Autophagy pathway: Cellular and molecular mechanisms - PMC
    This review will define this process and the cellular pathways required, from the formation of the double membrane to the fusion with lysosomes in molecular ...
  29. [29]
    Autophagy: process and function - Genes & Development
    Complete sequestration by the elongating phagophore results in formation of the autophagosome, which is typically a double-membraned organelle. This step is a ...
  30. [30]
    Molecules and their functions in autophagy - Nature
    Jan 19, 2012 · In this review, we summarize the molecular mechanisms of autophagy process, especially focusing on autophagosome formation.
  31. [31]
    An Overview of Autophagy: Morphology, Mechanism, and Regulation
    In mammalian cells, there are three primary types of autophagy: microautophagy, macroautophagy, and chaperone-mediated autophagy (CMA).
  32. [32]
    Autosis and autophagic cell death: the dark side of autophagy - Nature
    Sep 26, 2014 · Autosis is an autophagy-dependent non-apoptotic form of cell death, characterized by enhanced cell substrate adhesion, focal ballooning of the ...
  33. [33]
    Autosis as a selective type of cell death - Frontiers
    Autophagy enhances cell survival under many stress conditions, while in certain circumstances, autophagy promotes cell death (Liu and Levine, 2015).Introduction · Selectivity of autosis · Regulators of autosis · Future directions
  34. [34]
    Autophagy and Cell Death - PMC - PubMed Central - NIH
    Autophagy is intimately associated with eukaryotic cell death and apoptosis. Indeed in some cases the same proteins control both autophagy and apoptosis.
  35. [35]
    Necrosis Pathology - StatPearls - NCBI Bookshelf - NIH
    It is an uncontrolled cell death that results in swelling of the cell organelles, plasma membrane rupture and eventual lysis of the cell.
  36. [36]
    Necrotic death as a cell fate - Genes & Development
    In contrast, necrosis can be defined morphologically by electron-lucent cytoplasm, swelling of cellular organelles, and loss of plasma membrane integrity (Table ...Abstract · Physiological Roles And... · Energy Failure As A Cause Of...<|control11|><|separator|>
  37. [37]
    Histology, Cell Death - StatPearls - NCBI Bookshelf
    Jan 30, 2023 · It is usually the result of acute external factors, such as trauma, ischemia, toxins, or other non-physiologic mechanisms. ... influx of calcium ...
  38. [38]
    Necrosis - Pathologia - The University of Edinburgh
    Necrosis is the pattern of cell death that occurs in response to injuries such as hypoxia, extremes of temperature, toxins, physical trauma, and infection with ...
  39. [39]
    Is mPTP the gatekeeper for necrosis, apoptosis, or both? - PMC
    Recent observations suggest that mitochondrial permeabilization mediated by mPTP is generally more closely linked to events of late apoptosis and necrosis.
  40. [40]
    Mitochondrial Reactive Oxygen Species (ROS) and ROS-Induced ...
    At higher ROS levels, longer mPTP openings may release a ROS burst leading to destruction of mitochondria, and if propagated from mitochondrion to mitochondrion ...
  41. [41]
    Cell Death, Damage-Associated Molecular Patterns, and Sterile ...
    DAMPs released from necrotic cells ligate cognate or pattern recognition receptors leading to intracellular signaling, gene expression, and inflammation. Note ...
  42. [42]
    Apoptosis, Pyroptosis, and Necrosis: Mechanistic Description of ...
    This review presents a perspective on eukaryotic cell death and discusses experimental techniques used to study these processes.
  43. [43]
    Gasdermin D is an executor of pyroptosis and required for ... - Nature
    Nov 27, 2015 · GSDMD is required for pyroptosis and for the secretion but not proteolytic maturation of IL-1β in both canonical and non-canonical inflammasome responses.
  44. [44]
    Copper induces cell death by targeting lipoylated TCA cycle proteins
    Mar 17, 2022 · We show that copper-dependent death occurs by means of direct binding of copper to lipoylated components of the tricarboxylic acid (TCA) cycle.
  45. [45]
    Interdigital cell death in the embryonic limb is associated ... - Nature
    Sep 12, 2013 · Interdigital cell death is a physiological regression process responsible for sculpturing the digits in the embryonic vertebrate limb.
  46. [46]
    Programmed Cell Death in C. elegans - NCBI - NIH
    Most (113/131) developmental deaths occur during embryogenesis, mainly between 250 and 450 minutes after fertilization (Sulston et al. 1983). A second, smaller ...
  47. [47]
    Lens fibre cell differentiation and organelle loss: many paths lead to ...
    The programmed removal of organelles from differentiating lens fibre cells contributes towards lens transparency through formation of an organelle-free zone ...
  48. [48]
    Unconventional cell death in erythroid cells | Blood
    Jan 7, 2016 · Apoptosis and necroptosis are important mechanisms for the removal of diseased and damaged cells. Autophagy, which shares some features with ...
  49. [49]
    The role of apoptosis in the development and function of T ... - Nature
    Oct 1, 2005 · In the immune system, apoptosis is required for lymphocyte development and homeostasis. The dysregulation of apoptosis leads to a variety of ...
  50. [50]
    Autophagy in T‐cell differentiation, survival and memory - Wang - 2021
    Nov 3, 2020 · Autophagy maintains T cell development and survival by regulating the degradation of organelles and apoptotic proteins. The autophagic process ...Autophagy Modulates T Cell... · Autophagy Regulates T Cell... · Altered Autophagy In T Cells...
  51. [51]
    Necroptosis in anti-viral inflammation | Cell Death & Differentiation
    Jul 26, 2018 · Regulated necrosis, a proinflammatory cell death, potentially counteracts pathogenic infections ... viral clearance in response to VV ...
  52. [52]
    Cell death - ScienceDirect.com
    Jan 18, 2024 · Cell death supports morphogenesis during development and homeostasis after birth by removing damaged or obsolete cells.
  53. [53]
    The BCL-2 arbiters of apoptosis and their growing role as cancer ...
    Nov 3, 2017 · This review sketches the impact of the BCL-2 family on cancer development and therapy, describes how interactions of family members trigger apoptosis.Missing: paper | Show results with:paper
  54. [54]
    Bcl-2–family proteins and hematologic malignancies
    Apr 1, 2008 · Cloning of portions of the human BCL-2 gene was first reported in 1985 by Tsujimoto et al,20 who cloned the breakpoints from t(14;18) ...
  55. [55]
    Ferroptosis: the vulnerability within a cancer monster - JCI
    May 15, 2023 · As such, ferroptosis inducers, including GPX4 inhibitors, have some specificity in targeting cancer cells while sparing normal cells. Indeed, ...Abstract · Emerging roles of ferroptosis... · Ferroptosis inducers as...
  56. [56]
    The necroptosis cell death pathway drives neurodegeneration in ...
    Jun 9, 2024 · Recent evidence shows that necroptosis is abundant in AD, that necroptosis is closely linked to the appearance of Tau pathology, and that necroptosis markers ...
  57. [57]
    Regulated Cell Death in Myocardial Infarction | Circulation Research
    Aug 28, 2025 · Regulated cell death has been shown to contribute to the pathogenesis of the major cardiovascular syndromes, including atherosclerosis, ...
  58. [58]
    Pyroptosis in NLRP3 inflammasome-related atherosclerosis - PMC
    Oct 10, 2022 · Pyroptosis is a proinflammatory form of programmed cell death in response to inflammation. It involves in the pathogenesis and outcomes of atherosclerosis.
  59. [59]
    Pyroptosis in host defence against bacterial infection - PMC - NIH
    Summary: Pyroptosis is a form of inflammatory cell death. Here, we compare the interplay between pyroptosis and important human pathogens, i.e. Mycobacterium ...
  60. [60]
    To kill or be killed: viral evasion of apoptosis | Nature Immunology
    Nov 1, 2002 · In the struggle between virus and host, control over the cell's death machinery is crucial for survival.
  61. [61]
    BH3-mimetics: recent developments in cancer therapy
    Nov 9, 2021 · As seen in the context of most cancer-related diseases, the development of BH3-mimetics has been driven by them inducing cell death through ...Missing: cuproptosis | Show results with:cuproptosis
  62. [62]
    GPX4-independent ferroptosis—a new strategy in disease's therapy
    Oct 30, 2022 · These data suggest that DHODH inhibition induces ferroptosis in GPX4-low cancer cells while sensitizing GPX4-high cancer cells to ferroptosis.Nadph-Fsp1-Coq10 Pathway · P53-Mediated Lipoxygenase... · Ferritin And Prominin2
  63. [63]
    Cuproptosis: potential new direction in diabetes research and ...
    Jun 5, 2024 · Cuproptosis, a recently discovered form of cell death, stems from an overabundance of copper ions infiltrating mitochondria.
  64. [64]
    A fluorescent reporter of caspase activity for live imaging - PNAS
    Sep 16, 2008 · We have devised a genetically encoded caspase sensor that can be detected in live and fixed tissue by standard confocal microscopy.
  65. [65]
    Live imaging of apoptotic signaling flow using tunable combinatorial ...
    Dec 7, 2022 · Live imaging of apoptotic signaling flow using tunable combinatorial FRET-based bioprobes for cell population analysis of caspase cascades.Missing: seminal | Show results with:seminal
  66. [66]
    MLKL polymerization-induced lysosomal membrane ... - Nature
    Nov 23, 2023 · Our study demonstrates that upon induction of necroptosis, activated MLKL translocates to and polymerizes on the lysosomal membrane.
  67. [67]
    Mixed Lineage Kinase Domain‐Like Protein (MLKL): From ...
    Aug 11, 2025 · Han et al. used super-resolution microscopy to visualize necrosome and found that only RIPK3 oligomers with sizes of tetramer or above allow ...Mixed Lineage Kinase... · 2.3. 1 Ripk3-Mlkl Complex... · 2.4 Targeting Mlkl Inhibits...
  68. [68]
    CRISPR system for genome engineering: the application for ... - NIH
    Here, we reviewed the very recent study of the novel genome editing tool CRISPR in knockout of autophagy genes and reported our partial date in order to ...
  69. [69]
    CRISPR screening using an expanded toolkit of autophagy ...
    CRISPR screens for modifiers of autophagic flux recover known ATG factors and reveal new candidates. Next, we performed a series of genome-wide, pooled CRISPR ...
  70. [70]
    Cell-specific and athero-protective roles for RIPK3 in a murine ... - NIH
    This study reports that RIPK3 in macrophages and endothelial cells protects against atherosclerosis progression in mice.
  71. [71]
    Death-seq identifies regulators of cell death and senolytic therapies
    Sep 11, 2023 · Death-seq enables the systematic screening of cell death pathways to uncover molecular mechanisms of regulated cell death subroutines and ...<|separator|>
  72. [72]
    Proteome-wide identification of family member-specific ... - PNAS
    To identify natural family member specific caspase substrates on a proteome-wide scale, a systematic and broadly applicable strategy is greatly desired. mRNA ...
  73. [73]
    Caspases and their substrates - PMC - NIH
    May 12, 2017 · Significant progress has been made using modern proteomics methods to identify native protein substrates either in intact cells by triggering ...
  74. [74]
    Optogenetic apoptosis: light-triggered cell death - PubMed - NIH
    Oct 5, 2015 · An optogenetic Bax has been designed that facilitates light-induced apoptosis. We demonstrate that mitochondrial recruitment of a genetically encoded light- ...
  75. [75]
    Machine Learning Classifies Ferroptosis and Apoptosis Cell Death ...
    Mar 1, 2022 · TfR1 staining, combined with nuclear and F-actin staining, can reliably detect both apoptotic and ferroptotis cells when cell features are analyzed in an ...
  76. [76]
    Transcriptomic profiling of shed cells enables spatial mapping of ...
    Oct 2, 2025 · We characterize the turnover rates of diverse cell types and identify tissue microenvironments that may promote cell death. Results.Missing: 2020s | Show results with:2020s