Fact-checked by Grok 2 weeks ago

Caspase

Caspases are a family of cysteine-dependent aspartate-specific proteases that serve as critical regulators of , innate immunity, , and cellular in multicellular organisms. These enzymes are synthesized as inactive zymogens, consisting of a prodomain, a large catalytic subunit (17–20 kDa), and a small catalytic subunit (10–12 kDa), which are activated through proteolytic processing into an active heterotetramer. Humans express 12 caspases, while mice have 10, with their catalytic activity relying on a histidine-cysteine dyad that hydrolyzes bonds after residues. Caspases are broadly classified into three main groups based on function: inflammatory caspases (caspase-1, -4, and -5 in humans; caspase-11 in mice), which process pro-inflammatory cytokines like interleukin-1β (IL-1β) and interleukin-18 (IL-18) and trigger via gasdermin D cleavage; apoptotic initiator caspases (, -9, and -10), which activate downstream effectors in response to extrinsic death receptor signals or intrinsic mitochondrial stress; and apoptotic effector caspases (caspase-3, -6, and -7), which dismantle cellular structures by cleaving hundreds of substrates, including enzymes, cytoskeletal proteins, and nuclear lamins, leading to orderly cell demise without inflammation. Additional caspases, such as caspase-2, -10, -12, and -14, participate in non-apoptotic processes like cell differentiation, regulation, and tissue development. Beyond , caspases contribute to physiological processes, including embryonic development, immune cell activation, and remodeling, while dysregulation is implicated in diseases such as cancer, neurodegenerative disorders, and autoimmune conditions, making them promising therapeutic targets. The first caspase (caspase-1, originally termed interleukin-1β converting enzyme) was cloned in 1992, and its structure was solved in 1994, marking the beginning of extensive research into their mechanisms.

Overview and Structure

Definition and Properties

Caspases constitute a family of cysteine-dependent aspartate-specific proteases, commonly referred to as cysteine aspartases, that selectively cleave peptide bonds C-terminal to aspartic acid residues in their protein substrates. This enzymatic specificity distinguishes them from other protease families and is central to their biological roles. Caspases are evolutionarily conserved across metazoans, with homologs identified in organisms ranging from nematodes like Caenorhabditis elegans to mammals, underscoring their fundamental importance in multicellular life. These proteases are synthesized as inactive precursors known as zymogens or procaspases, which must undergo to exert their catalytic function. typically involves proteolytic processing, ensuring tight to prevent untimely cellular damage. A hallmark of their structure is the conserved catalytic dyad composed of a residue and a residue within the , which facilitates efficient . In humans, 12 distinct caspases have been identified, reflecting their diverse yet coordinated contributions to cellular processes. The catalytic mechanism of caspases relies on the nucleophilic attack by the group of the active-site on the carbonyl carbon of the , with the adjacent acting as a general base to deprotonate the and enhance its reactivity. This process exhibits stringent specificity for at the P1 position of the , a feature that ensures precise targeting of caspase substrates during cellular signaling. Caspases play essential roles in maintaining and overall cellular , processes critical for development, tissue remodeling, and immune responses in metazoans.

Molecular Architecture

Caspases are synthesized as inactive zymogens known as procaspases, which consist of an N-terminal prodomain of variable length, followed by a large catalytic subunit (approximately 20 , or p20) and a small catalytic subunit (approximately 10 , or p10). The prodomain, typically 20–120 long, is absent or short in effector caspases but extended in initiator caspases to include specific interaction motifs such as the caspase recruitment (CARD) or death effector (DED), which facilitate oligomerization and signaling. These domains enable homotypic interactions with adaptor proteins in platforms like the or . In their mature, active form, caspases assemble into a heterotetrameric structure comprising two p20 subunits and two p10 subunits, often denoted as (p20)₂(p10)₂. The active sites are located at the interfaces between the p20 and p10 subunits of adjacent monomers, where loops from both subunits contribute to recognition and . This quaternary arrangement ensures that the enzyme's proteolytic activity is tightly regulated and only manifests upon proper dimerization. The catalytic domains of caspases exhibit a conserved beta-sheet-rich fold, featuring a central twisted six-stranded beta-sheet flanked by five alpha-helices on one side and additional helices and loops on the other, forming a compact globular structure. At the heart of the active site lies the highly conserved QACRG pentapeptide motif, where the cysteine residue (C) serves as the nucleophilic attacker in peptide bond hydrolysis, and the arginine (R) helps position the substrate. Caspases also harbor allosteric sites, often at dimer interfaces or remote loops, that modulate activity by stabilizing inactive conformations or enhancing catalytic efficiency upon ligand binding. Activation involves proteolytic processing that removes the prodomain and intersubunit linkers, inducing significant conformational changes, particularly in the loops (L1–L4). These rearrangements expose the substrate-binding cleft, which is occluded in the procaspase form, thereby enabling access for aspartate-containing substrates and full enzymatic competence. Dimerization further stabilizes this open conformation, linking architecture to function.

Classification

Initiator and Effector Types

Caspases are classified into initiator and effector types based on their structural features, activation mechanisms, and sequential roles within proteolytic cascades that drive . This reflects their positions in signaling pathways, where initiators serve as sensors and amplifiers at the , while effectors act downstream to dismantle cellular components. In humans, twelve caspases are identified (numbered 1 through 10, 12, and 14), with most fitting neatly into these categories, though caspase-2 represents an atypical case that can function in both capacities depending on context. Initiator caspases, including , , and caspase-10, are characterized by long N-terminal prodomains that facilitate their recruitment to platforms. These prodomains typically contain death effector domains (DEDs) in caspase-8 and -10 or a caspase-recruitment domain (CARD) in caspase-9, enabling oligomerization and auto- through induced proximity upon receptor or sensor engagement. As the starting points of caspase cascades, initiator caspases amplify upstream death signals by undergoing conformational changes that enhance their catalytic activity, thereby processing downstream targets to propagate the response. In contrast, effector caspases, such as caspase-3, caspase-6, and caspase-7, feature short prodomains and exist predominantly as inactive zymogens until cleaved by active initiator caspases. This proteolytic activation removes the prodomain and separates the large and small subunits, forming the mature heterotetramer that executes the terminal phase of the . Effector caspases specialize in the bulk of cellular substrates, including structural proteins and regulatory factors, leading to systematic disassembly of the cell. The model underscores this functional hierarchy: initiator caspases detect and integrate apoptotic stimuli to initiate signal amplification, while effector caspases, once activated, perform the widespread that commits the to . Caspase-2, with its long CARD-containing prodomain, blurs these lines as an member capable of initiator-like auto-activation in some scenarios or effector-like roles in others, highlighting the nuanced diversity within the family.

Inflammatory Subtypes

Inflammatory caspases constitute a specialized subclass of the caspase family, distinct from those primarily involved in , and are dedicated to orchestrating innate immune responses through maturation and, in some cases, lytic . In humans, this group encompasses caspase-1, caspase-4, and caspase-5, while mice express caspase-11 as the ortholog to human caspase-4 and -5. These enzymes are characterized by an N-terminal caspase recruitment domain (), which enables their assembly into multiprotein complexes, facilitating proximity-induced activation via dimerization and autoproteolytic cleavage. Caspase-1 serves as the canonical inflammatory caspase, recruited to such as or AIM2, where it selectively cleaves pro-IL-1β and pro-IL-18 at specific aspartate residues (Asp116 for IL-1β and Asp36 for IL-18), yielding mature that amplify . Beyond cytokine processing, caspase-1 also targets gasdermin D (GSDMD) at Asp276, generating an N-terminal fragment that forms plasma membrane pores, which promotes IL-1 family cytokine release and executes —a proinflammatory form of . In contrast, human caspase-4 and -5, along with murine caspase-11, operate in non-canonical pathways, directly binding (LPS) via their domains to sense cytosolic Gram-negative bacterial components; this leads to GSDMD cleavage and , often indirectly enhancing caspase-1 activity for cytokine production. These caspases exhibit substrate specificity favoring inflammatory mediators over the structural proteins dismantled by apoptotic executioners like caspase-3 and -7. Post-2020 research has illuminated expanded regulatory roles for these caspases within the , highlighting mechanisms such as NEK7-mediated oligomerization for caspase-1 and cross-talk with non-canonical sensors like caspase-11 to integrate diverse danger signals. Structural studies have revealed how CARD-CARD interactions form filamentous assemblies, providing targets for therapeutic modulation in inflammatory diseases. These findings underscore the inflammatory caspases' pivotal position in bridging microbial detection to adaptive immune amplification, distinct from their apoptotic counterparts' focus on orderly cell demise.

Activation Mechanisms

Dimerization Processes

Caspase activation frequently initiates through the dimerization of inactive procaspase monomers, a process that stabilizes the active conformation and facilitates subsequent proteolytic events. In this , homodimerization of the domains brings the catalytic sites into proximity, enabling low-level autoproteolytic activity that is amplified upon clustering. This step is essential for initiator caspases, such as and , where the monomeric zymogens exhibit negligible activity due to disordered loops, but dimerization induces conformational changes that align these loops for . Adaptor proteins or scaffold complexes induce dimerization by promoting the oligomerization of procaspases, thereby increasing their local concentration and overcoming the intrinsically low affinity for self-association. For , recruitment to the (), formed by () and death receptors like /CD95, drives homodimerization through homophilic interactions between the domains. Similarly, dimerizes upon binding to the , a heptameric platform assembled from apoptotic protease-activating factor 1 (Apaf-1) and released from mitochondria. These inducers create a high-density environment that shifts the equilibrium toward the dimeric state, essential for initiating the caspase cascade. The kinetics of dimerization follow the induced proximity model, in which adapter-mediated clustering compensates for the weak dimerization affinity (typically in the micromolar range) by elevating effective concentrations to nanomolar levels within the complex. This proximity-driven process results in a second-order for , as demonstrated for on the , where monomer-dimer equilibrium favors inactivity at physiological concentrations but shifts rapidly upon scaffolding. For , interdimer processing within DISC oligomers further accelerates , with dimers cross-cleaving each other to expose processing sites. This model explains the rapid onset of activity without requiring high-affinity intrinsic interactions.00173-0) Structurally, the dimer interface involves conserved residues from β-strands and α-helices that mediate subunit contacts, allosterically regulating the . In , key interactions occur at a seven-residue insertion loop between β-strands 3 and 3A (position 240), which spans the dimer interface and stabilizes the L1 and loops critical for binding and . Caspase-8 employs similar homophilic contacts in its protease domain, with residues in the death effector domains (DEDs) contributing to stable dimer formation upon assembly. These interface residues not only promote dimer stability but also induce allosteric rearrangements that enhance catalytic efficiency by up to 100-fold compared to monomers. This structural basis underscores dimerization as a regulatory switch for caspase activation.

Proteolytic Cleavage

Caspases are synthesized as inactive zymogens known as procaspases, which require proteolytic to generate mature, active enzymes. This maturation involves at specific residues within the protein sequence, primarily following the consensus motif DXXD↓, where the arrow indicates the cleavage site after the terminal aspartate. These cleavage events occur in interdomain linkers: between the N-terminal prodomain and the large subunit (p20), and between the large and small subunits (p10), resulting in the heterotetrameric active form composed of two large and two small subunits. The proteolytic cascade amplifies the apoptotic signal through a hierarchical process. Initiator procaspases, such as and , undergo initial auto-cleavage or trans-cleavage upon dimerization induced by upstream adaptors, generating partially active forms that then proteolytically process effector procaspases like caspase-3 and caspase-7. Effector caspases require cleavage at multiple sites for full activation but exhibit minimal auto-processing capability, relying predominantly on initiator caspases for their maturation; once activated, effectors further cleave downstream substrates to execute . This sequential amplification ensures rapid and irreversible commitment to . Regulation of proteolytic cleavage is tightly controlled by endogenous inhibitors, notably the X-linked inhibitor of apoptosis protein (XIAP). XIAP binds directly to the active sites of processed effector caspases (caspase-3 and -7) and the linker region of initiator , preventing substrate access and further cleavage events. Relief of this inhibition occurs through mitochondrial release of second mitochondria-derived activator of caspases (Smac/DIABLO), which binds XIAP's BIR domains, displacing caspases and allowing progression of the proteolytic cascade. Recent studies have highlighted non-canonical proteolytic mechanisms in inflammatory contexts, particularly involving inflammatory caspases such as caspase-1, -4, -5, and -11. These caspases can cleave substrates like gasdermin D (GSDMD) at atypical sites deviating from the strict DXXD motif, facilitating without traditional priming; for instance, caspase-11 autoprocessing enables direct recognition of bacterial , leading to GSDMD pore formation and IL-1β/IL-18 processing. Such insights from 2024 analyses underscore the adaptability of caspase cleavage in innate immunity beyond canonical .

Roles in Programmed Cell Death

Apoptosis Pathways

Apoptosis, a form of programmed cell death essential for development and homeostasis, is primarily executed through caspase-mediated proteolysis that systematically dismantles cellular structures. Caspases serve as central effectors in two major pathways: the extrinsic pathway, triggered by extracellular signals, and the intrinsic pathway, initiated by internal stresses such as DNA damage or mitochondrial dysfunction. In both, initiator caspases activate effector caspases, leading to irreversible cellular commitment to death while ensuring orderly fragmentation for efficient clearance by phagocytes. The extrinsic pathway is activated by ligand binding to death receptors, such as (CD95) or (TNFR1), recruiting the adaptor protein and initiator to form the death-inducing signaling complex (). Caspase-8 undergoes dimerization and autocleavage, becoming active and directly cleaving effector caspases like caspase-3 in type I cells, or indirectly amplifying the signal in type II cells by cleaving Bid to generate truncated Bid (tBid). tBid translocates to mitochondria, promoting Bax/Bak oligomerization and release, thereby linking the extrinsic pathway to the intrinsic amplification loop. In the intrinsic pathway, mitochondrial outer membrane permeabilization releases into the , where it binds Apaf-1 in the presence of dATP, inducing Apaf-1 oligomerization into a wheel-like heptameric complex. This structure recruits and activates initiator through induced proximity and autocleavage, with remaining partially associated with the to process effector caspases such as caspase-3 and -7. The pathway integrates diverse stressors, including genotoxic damage and deprivation, ensuring a robust response to cellular threats. Effector caspases, primarily caspase-3 and caspase-7, then cleave over 200 substrates to orchestrate apoptotic morphology and function. Caspase-3 cleaves poly(ADP-ribose) polymerase (PARP) at Asp214, abrogating DNA repair and conserving energy for dismantling; caspase-6 targets nuclear lamins A/C at Asp230, fragmenting the nuclear envelope to enable chromatin condensation. DNA fragmentation occurs via caspase-3-mediated cleavage of inhibitor of caspase-activated DNase (ICAD) at Asp117 and Asp224, liberating CAD to generate 180-bp nucleosomal ladders. Cytoskeletal changes drive membrane blebbing through caspase-3 cleavage of gelsolin at Asp352, promoting actin depolymerization, and Rho-associated kinase 1 (ROCK1) at Asp1113, enhancing actomyosin contraction. Phagocytosis is facilitated by caspase-7 cleavage of endonuclease G-like 1 (EMAP-II) and lipid scramblases like ATP11C at Asp439, exposing phosphatidylserine on the outer membrane to signal immune clearance without inflammation. Beyond full commitment to death, caspases can operate at sublethal thresholds, enabling partial for cellular remodeling without complete . Low-level caspase-3 activity, for instance, supports in megakaryocytes and myoblasts by cleaving select substrates like regulators, facilitating morphological changes such as adjustments during terminal maturation. This regulated, compartmentalized avoids widespread substrate degradation, highlighting caspases' dual roles in both demise and .

Pyroptosis Mechanisms

Pyroptosis is a form of programmed lytic characterized by the formation of pores, leading to osmotic cell and the release of pro-inflammatory contents, primarily mediated by inflammatory caspases such as caspase-1, -4, -5, and -11 acting on gasdermin D (GSDMD). This process serves as a host defense mechanism against microbial infections but can contribute to pathological when dysregulated. Unlike , pyroptosis amplifies immune responses through secretion and cell rupture. In the canonical pathway, pattern recognition receptors like or AIM2 detect pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs), leading to assembly with the adaptor protein ASC and pro-caspase-1. This complex autocleaves pro-caspase-1 into its active form, which then proteolytically cleaves GSDMD at Asp275 (in humans), separating the N-terminal fragment (GSDMD-N) from the inhibitory C-terminal domain. The GSDMD-N domain oligomerizes and inserts into the plasma membrane, forming pores approximately 10-20 nm in diameter that disrupt membrane integrity. Concurrently, active caspase-1 matures pro-interleukin-1β (pro-IL-1β) and pro-IL-18 into their bioactive forms, which are released through these pores to propagate . The non-canonical pathway operates independently of and is triggered by direct sensing of bacterial (LPS) in the by caspase-4 and -5 in humans or caspase-11 in mice. These caspases bind LPS via their domains, undergo autoprocessing, and cleave GSDMD at the same site as caspase-1, releasing GSDMD-N to form pores. Pore formation induces potassium efflux, which secondarily activates the and caspase-1, amplifying IL-1β and IL-18 release. This pathway is particularly relevant in Gram-negative bacterial infections, where it promotes rapid pyroptotic responses. The outcomes of both pathways include cell swelling, membrane rupture, and the extracellular release of inflammatory mediators, fostering immune cell recruitment and amplifying systemic inflammation. In recent studies, hyperactivation of pyroptosis via NLRP3 inflammasome and GSDMD has been implicated in metabolic diseases; for instance, in type 2 diabetes, excessive caspase-1-mediated GSDMD cleavage in pancreatic β-cells exacerbates insulin resistance and β-cell dysfunction. Similarly, in obesity and metabolic-associated fatty liver disease, adipocyte and hepatocyte pyroptosis drives chronic low-grade inflammation, linking pyroptosis to disease progression. These findings highlight pyroptosis as a potential therapeutic target in metabolic disorders.

Involvement in Necroptosis and Ferroptosis

Caspases, particularly , play a critical regulatory role in necroptosis by acting as inhibitors of the RIPK3-MLKL signaling pathway. In this lytic form of , caspase-8 normally cleaves receptor-interacting protein kinase 1 () at aspartate 325 (in humans) and RIPK3 at aspartate 328, thereby disrupting the formation of the -RIPK3 necrosome complex that phosphorylates and activates mixed lineage kinase domain-like protein (MLKL). When caspase-8 is inhibited or absent, such as through viral infection or pharmacological blockade, RIPK1 and RIPK3 evade cleavage, undergo , and assemble the necrosome, leading to MLKL oligomerization, translocation, and subsequent rupture characteristic of necroptotic cell lysis. This inhibitory function positions caspase-8 as a , preventing necroptosis under conditions where apoptotic signaling predominates. In , an iron-dependent driven by unchecked , caspases do not directly execute the process but exert indirect regulatory influence, with caspase-3 notably promoting susceptibility through modulation of lipid peroxidation regulators. Activation of caspase-3, often triggered by apoptotic stimuli, can sensitize cells to ferroptotic execution, shifting the balance toward ferroptotic outcomes in stressed cells without direct involvement in the core peroxidation cascade. Unlike necroptosis, where caspase inhibition unleashes the pathway, ferroptosis proceeds independently of caspases, but caspase-3's proteolytic activity indirectly amplifies lipid peroxidation in contexts where apoptotic pathways are engaged but incomplete. This non-executory role underscores caspases' broader influence on alternative death modalities when primary apoptotic pathways are engaged but incomplete. Crosstalk between caspase-mediated apoptosis and these alternative deaths emerges prominently when apoptotic execution is suppressed, redirecting cells toward necroptosis or in contexts like cancer and . In tumor microenvironments, inhibition of allows RIPK3-MLKL activation, promoting necroptotic that enhances antitumor immunity, while in viral infections, caspase blockade similarly shifts to necroptosis for pathogen clearance. Similarly, apoptosis-resistant cancer cells, where caspase-3 activity is dampened, default to upon inducers, providing a therapeutic exploited in infections where microbial evasion of caspases triggers ferroptotic . This compensatory switching ensures cell elimination despite pathway blockade, with caspases serving as pivotal integrators. Recent reviews highlight caspases' potential in sensitizing ferroptosis for cancer therapy, particularly through combined modulation of apoptotic and ferroptotic axes to overcome resistance. For instance, caspase-3 activation via BH3-mimetics enhances lipid peroxidation in apoptosis-refractory tumors, amplifying ferroptosis inducers like RSL3 for synergistic cell killing. In 2025 analyses, caspase inhibition strategies are proposed to redirect suppressed apoptosis toward ferroptosis in immunotherapy-resistant cancers, improving outcomes in infection-associated malignancies by leveraging necroptotic-ferroptotic crosstalk. These findings emphasize caspases' therapeutic targeting to fine-tune cell death switches for enhanced efficacy.

Non-Apoptotic Functions

Regulation of Inflammation

Caspases, particularly caspase-1, play a central role in regulating by processing inactive precursor cytokines into their mature, bioactive forms, thereby amplifying innate immune responses. Caspase-1 specifically cleaves pro-interleukin-1β (pro-IL-1β) at the Asp116-Ala117 bond to generate the mature 17 kDa IL-1β protein, which acts as a potent mediator of fever, endothelial activation, and recruitment of immune cells. Similarly, caspase-1 processes pro-IL-18 into its 17.2 kDa mature form, promoting interferon-γ production and enhancing Th1 immune responses. These maturation events are essential for the cytokines' and , enabling rapid propagation of inflammatory signals without requiring de novo protein synthesis. Inflammasomes serve as critical platforms integrating caspase activation with pathogen and damage sensing to fine-tune inflammatory outputs. Nucleotide-binding oligomerization domain-like receptors (NLRs), such as , detect pathogen-associated molecular patterns (PAMPs) from microbes or damage-associated molecular patterns (DAMPs) from host cells, leading to NLR oligomerization. This recruits the adaptor protein apoptosis-associated speck-like protein containing a (ASC), which facilitates proximity-induced autocleavage and activation of pro-caspase-1 into its enzymatically active p20/p10 heterotetramer form. The resulting active caspase-1 then drives maturation as a primary signaling mechanism, distinct from other effector functions. Caspase activation establishes positive feedback loops that sustain pro-inflammatory signaling through crosstalk with transcription factors like . Mature IL-1β and IL-18 secreted downstream of caspase-1 bind to their respective receptors (IL-1R and IL-18R), triggering intracellular cascades that activate the (IKK) complex and liberate from inhibitory sequestration. Nuclear translocation of then induces transcription of additional pro-inflammatory genes, including those encoding more pro-IL-1β, thereby amplifying and prolonging the inflammatory response in an autocrine and paracrine manner. This loop ensures robust innate immunity but requires tight regulation to prevent chronic inflammation. Dysregulated caspase overactivation contributes to pathological inflammation in conditions like and , where unchecked release exacerbates tissue damage. In , sustained inflammasome-caspase-1 signaling, often via or NLRC4, drives excessive IL-1β production, fueling a systemic that correlates with multi-organ failure and high mortality rates exceeding 20% in severe cases. Recent analyses indicate that inhibiting this pathway reduces inflammatory burden in preclinical models. In , infection hyperactivates inflammasomes, leading to elevated caspase-1 activity and IL-1β/IL-18 levels in severe patients, with studies showing caspase-1 activation in lung biopsies, linking this to immune dysregulation. Therapeutic targeting of caspase-1 has shown promise in mitigating these effects.

Roles in Differentiation and Stem Cells

Caspases, particularly caspase-3, exhibit sublethal activation that supports non-lethal cellular processes during and . In neuronal , low-level caspase-3 activity facilitates synapse pruning by enabling activity-dependent elimination of synaptic connections, a process essential for circuit refinement without inducing . This sublethal function extends to promoting neuronal migration, where caspase-3 enhances cytoskeletal dynamics and cell motility in non-apoptotic contexts, aiding proper positioning in the developing . In stem cell biology, caspase-3 plays a regulatory role in maintaining () quiescence, where its partial activation dampens responsiveness to extrinsic signals, thereby preserving the long-term repopulating potential of these cells. These non-apoptotic activities highlight caspase-3's contribution to durability and tissue regeneration. During , caspases cleave key s to drive lineage commitment. In lens development, executioner caspases, including caspase-3, are activated in fiber cells to degrade nuclear components and facilitate differentiation, with caspase activity targeting regulatory proteins for precise organelle elimination. Similarly, in , caspase-3 cleaves the Pax7 in satellite cells, inhibiting self-renewal and promoting progression to myogenic by releasing cells from a stem-like state. Recent insights as of 2025 underscore caspase-3's involvement in (ESC) pluripotency, where sublethal activation cleaves Nanog to initiate while preserving cell viability, linking partial caspase signaling to the exit from naive pluripotency. In cancer stem cells, non-genetic inactivation of caspase-3 enhances survival and stemness, allowing tumor-initiating cells to evade and promote resistance to therapies through sustained low-level activity that supports and . These findings emphasize caspase-3's paradoxical role in balancing survival and fate decisions in stem-like populations.

Historical Development

Discovery and Initial Characterization

In the 1980s, genetic screens conducted in the nematode by and colleagues identified the gene ced-3 as essential for (apoptosis) during development. Mutations in ced-3 blocked nearly all cell deaths that occur in the wild-type worm, revealing it as a key executor of the apoptotic program. This discovery established ced-3 as a central component of the genetically defined apoptotic pathway in , alongside genes like ced-4 and ced-9. In 1993, the ced-3 gene was cloned and found to encode a protein highly similar to the human interleukin-1β (IL-1β) converting (ICE), previously and cloned in 1992 as a responsible for processing the IL-1β precursor into its mature form. The ced-3 product shared about 30% sequence identity with ICE (now known as caspase-1), suggesting that ICE represented a mammalian homolog involved in . Overexpression of either ced-3 or ICE in mammalian fibroblasts induced apoptotic , linking the two proteins functionally across . Early characterization of these proteases relied on cell-free systems, particularly reconstitution assays using extracts from laevis eggs, which recapitulated key apoptotic events such as nuclear fragmentation and DNA laddering. These extracts demonstrated caspase-like proteolytic activity essential for , allowing researchers to purify and study the enzymes' substrate specificities and activation mechanisms without the complexities of intact cells. The unified nomenclature "caspase" (for cysteine-dependent aspartate-specific proteases) was proposed in by an international committee to encompass , CED-3, and related family members, standardizing the naming of this growing class of regulators.

Key Milestones and Advances

In 1997, the identification of the as a key activation platform for marked a pivotal advance in understanding the intrinsic pathway of . This multiprotein complex, formed by Apaf-1 oligomerization in the presence of and dATP, recruits and activates procaspase-9, initiating the downstream caspase cascade that executes . The discovery elucidated how mitochondrial signals converge to amplify caspase activity, providing a mechanistic foundation for caspase regulation in apoptotic signaling. During the 2000s, the discovery of the complex revolutionized insights into caspase-1's role beyond , particularly in and innate immunity. In 2002, researchers identified the as a molecular platform comprising NLR family proteins, ASC adaptor, and pro-caspase-1, which oligomerizes in response to microbial or danger signals to process pro-caspase-1 into its active form. This activation cleaves gasdermin D and pro-IL-1β, driving release and lytic in , thereby linking caspases to host defense against pathogens. Subsequent studies in the decade expanded this to multiple inflammasome sensors, highlighting caspase-1's central role in inflammation-related pathologies. The 2010s unveiled extensive non-apoptotic functions of caspases, particularly in biology and , broadening their physiological repertoire. For instance, low-level caspase-3 activity was shown to promote epigenetic and pluripotency in induced pluripotent s (iPSCs) without inducing . This revealed caspases as regulators of maintenance, proliferation, and lineage commitment in tissues like the hematopoietic system and neural progenitors, where partial activation supports rather than demise. These findings shifted paradigms, positioning caspases as versatile proteases in development and regeneration. In 2024, advances in cryo-electron microscopy (cryo-EM) provided high-resolution structures of caspase-involved complexes, enhancing therapeutic targeting for autoimmune diseases. Cryo-EM revealed detailed oligomerization states of bound to ASC and , illustrating nucleotide-dependent activation mechanisms that drive in inflammatory contexts. Concurrently, preclinical studies of inhibitors like VX-765 for inflammatory and autoimmune conditions, such as , have demonstrated reduced IL-1β production and inflammation in disease models by blocking inflammasome-driven storms.

Evolutionary Aspects

Origins and Conservation

Caspases belong to a superfamily of proteases that traces its origins to early eukaryotic , with metacaspases identified in and fungi as distant relatives. These metacaspases, such as those in and , share a common ancestral fold with animal caspases but exhibit distinct substrate preferences. Paracaspases, another branch of this superfamily, are present in pre-metazoan organisms such as Dictyostelium discoideum, while metacaspases are found in choanoflagellates, the closest unicellular relatives to , indicating that caspase-like machinery evolved prior to the advent of multicellularity in the metazoan . A key conserved feature across the caspase superfamily is the catalytic dyad composed of a and residue, which is preserved from metacaspases like Yca1 to human caspases, enabling nucleophilic attack during . The (Asp) specificity at the P1 position of substrates, however, is a hallmark unique to animal caspases and remains highly conserved from early metazoans to humans, distinguishing them from metacaspases that prefer basic residues like . This structural conservation highlights the fundamental mechanistic similarity despite functional divergence. In the lineage, the underwent significant expansion through tandem and segmental duplications, resulting in specialized isoforms for , , and non-death functions. For instance, early genome duplications contributed to the diversification of initiator and effector caspases, with s possessing around 12 active members compared to fewer in , allowing fine-tuned responses to cellular signals. A 2025 study highlighted the of caspase-16, where its prodomain arose from duplication of the caspase domain, with the ortholog rendered a by a . The primordial function of these proteases likely centered on stress responses in unicellular eukaryotes, as evidenced by metacaspases in choanoflagellates and yeast mediating adaptation to oxidative and environmental stresses through lateral gene transfer from algae, predating their co-option into metazoan apoptosis pathways.

Phylogenetic Distribution

Caspases, as cysteine-dependent aspartate-specific proteases, are exclusively found in metazoans, while their homologs known as metacaspases are distributed in non-metazoan eukaryotes such as plants and fungi. In plants, metacaspases contribute to hypersensitive response-like cell death during pathogen defense, with multiple isoforms identified across species like Arabidopsis thaliana. Fungi also possess metacaspases, which are involved in developmental and stress-related processes, reflecting a broad presence in the fungal kingdom. True metacaspases and caspases are absent in bacteria, though scattered prokaryotic homologs with caspase-like domains exist in some lineages, indicating independent evolutionary origins rather than direct ancestry. Within metazoans, the caspase family is present across phyla, with the full complement of initiator, effector, and inflammatory subtypes evident in bilaterians such as arthropods, nematodes, and chordates. In contrast, non-bilaterian metazoans like cnidarians exhibit a caspase repertoire that is less diverse in terms of subgroup specialization, with genomes such as that of magnipapillata encoding around 15 genes but lacking certain bilaterian-specific expansions in apoptotic initiators. This reduction in cnidarian caspases highlights adaptations to simpler body plans, while core catalytic domains remain conserved from the metazoan ancestor. In vertebrates, the caspase family has undergone significant expansion through gene duplications, resulting in 12 distinct genes in humans (CASP1 through CASP10 and CASP14), which arose from tandem and whole-genome duplication events in early vertebrate . Recent phylogenetic analyses in have revealed clade-specific innovations in inflammatory caspases, such as primate-specific duplications and sequence divergences in CASP1 and related genes, enabling specialized roles in innate immunity. These findings underscore the dynamic of caspases across phylogenetic clades, building on their conserved ancestral features.

Detection and Analysis

Biochemical Assays

Biochemical assays for caspases primarily involve and measurements of proteolytic activity using synthetic peptide substrates that mimic natural cleavage sites. These methods quantify through the release of detectable reporter groups, enabling precise evaluation of activation, inhibition, and specificity in recombinant proteins or cellular extracts.39097-5/fulltext) Fluorogenic substrates are widely used due to their sensitivity in detecting low levels of caspase activity. For instance, Ac-DEVD-AMC serves as a selective substrate for caspase-3, where cleavage at the residue liberates 7-amino-4-methylcoumarin (), producing with at approximately 380 nm and at 460 nm.39097-5/fulltext) Similarly, Ac-YVAD-AMC is specific for caspase-1, undergoing hydrolysis to release fluorescent under analogous conditions, allowing monitoring of to determine reaction rates via Michaelis-Menten parameters such as values around 10-20 μM for these substrates.39097-5/fulltext) These assays are typically performed in 96-well plates with lysates or purified enzymes, providing linear fluorescence increases over time that correlate with caspase concentration. Caspase activity assays often employ recombinant enzymes or lysates from treated cells to assess activation and inhibitor efficacy. In these setups, substrates like DEVD-AMC are incubated with samples, and activity is quantified by the rate of release, often normalized to protein content. Inhibitors such as z-VAD-fmk, a cell-permeable pan-caspase fluoromethylketone, are evaluated for potency, with values typically in the low nanomolar to micromolar range (e.g., 1-50 nM for caspase-3), determined by dose-response curves monitoring substrate cleavage inhibition. These assays confirm specificity and are essential for validating caspase involvement in apoptotic pathways using controls like untreated lysates. Specificity profiling of caspases utilizes positional scanning synthetic combinatorial libraries (PS-SCLs) to identify optimal substrate motifs. These libraries systematically vary at P4-P1 positions upstream of the , revealing preferences such as for caspases like caspase-3 and YVAD/WEHD for initiator caspases like caspase-1.39097-5/fulltext) By measuring cleavage rates of library subsets with fluorogenic reporters, researchers derive sequences that guide design and distinguish caspase subtypes based on extended subsite requirements.39097-5/fulltext) High-throughput screening adaptations enhance these assays for , incorporating fluorescence resonance energy transfer ()-based probes or luminescent detection. substrates, such as those linking donor-acceptor fluorophores across a caspase-cleavable linker (e.g., ), exhibit decreased energy transfer and increased donor emission upon , enabling 384-well format readouts with Z' factors >0.7 for robust screening. Luminescent assays, like the Caspase-Glo system, use proluminogenic analogs that generate via upon cleavage, offering superior signal-to-noise ratios (>100-fold) and automation compatibility for evaluating thousands of compounds against caspase activity in lysates.

Imaging Techniques

Fluorescent probes have been instrumental in visualizing caspase activation at the cellular level, particularly for effector caspases like caspase-3. PhiPhiLux represents a prominent class of cell-permeable fluorogenic substrates designed specifically for caspase-3 and -7 detection; upon by active caspase-3, the probe releases a fluorescent moiety, such as in the green-emitting G1D2 or red-emitting G2D2 variants, enabling of in live cells via confocal or widefield . These probes offer high specificity and low background fluorescence, allowing localization of caspase activity within subcellular compartments like the or . Activatable cell-penetrating (ACPPs) extend this capability by incorporating a polycationic cell-penetrating masked by a caspase-cleavable linker, which, upon activation, permits entry into cells and subsequent or near-infrared signal generation. For instance, the KcapQ probe, an improved near-infrared fluorescent ACPP, targets effector caspase activity in whole cells and live animals, providing enhanced signal-to-noise ratios for non-invasive imaging compared to non-activatable counterparts. This approach has been particularly useful in tracking caspase-mediated processes in tissues with limited accessibility, such as the or . Förster resonance energy transfer (FRET)-based s provide dynamic, real-time monitoring of caspase through intramolecular quenching that is relieved upon . The SCAT3 , specific for caspase-3, consists of enhanced fluorescent protein (ECFP) and (YFP) flanking a cleavage sequence; in the intact state, FRET occurs between the donor (ECFP) and acceptor (YFP), but caspase-3 cleavage separates them, shifting emission from to for ratiometric of in living cells. This has revealed spatial and temporal heterogeneity in caspase-3 onset during , with applications in high-resolution confocal setups to map within cellular structures. In vivo imaging techniques extend caspase visualization to whole organisms, facilitating longitudinal studies of and . Bioluminescent reporters, such as those fusing to a caspase-3-cleavable linker (e.g., pro-luciferin-DEVD), enable non-invasive detection in models; substrate administration leads to upon , quantifying caspase activity in tissues like tumors or inflamed organs with high sensitivity. For instance, this approach has monitored caspase-3 in hepatic apoptosis following hydrodynamic gene delivery, correlating bioluminescence intensity with disease progression. (PET) tracers target caspase activation in inflammatory contexts, with [18F]ICMT-11 serving as a selective inhibitor-based probe for caspase-3/7; it accumulates in apoptotic cells via irreversible binding, allowing quantification of in murine models of acute or tumors. Recent developments include 18F-labeled activity-based probes that enhance specificity for response in vivo. Advances as of 2025 have refined the spatial resolution of caspase-related structures, particularly the , through live-cell imaging techniques. Studies employing GFP-tagged Apaf-1 in human cell lines have demonstrated that apoptosomes form as large, transient assemblies of Apaf-1 oligomers upon cytochrome c release, visualized via time-lapse to capture their dynamic assembly and disassembly during mitochondrial . While super-resolution methods like STED have been applied to related apoptotic complexes, ongoing integration with these approaches promises sub-diffraction limit insights into recruitment and activation on the apoptosome platform. These techniques complement biochemical assays by providing spatial context for caspase localization without disrupting cellular integrity.

References

  1. [1]
    Caspases: structural and molecular mechanisms and functions in ...
    May 5, 2025 · Caspases are critical regulators of cell death, development, innate immunity, host defense, and disease.
  2. [2]
    Proteases for Cell Suicide: Functions and Regulation of Caspases
    Each caspase is a cysteine aspartase; it employs a nucleophilic cysteine in its active site to cleave aspartic acid peptide bonds within proteins. Caspases are ...
  3. [3]
    Caspases: pharmacological manipulation of cell death - PMC
    The CARD, the DED, and the large (p20) and small (p10) catalytic subunits are indicated. (B) Scheme of procaspase activation. Cleavage of the procaspase at the ...
  4. [4]
    The protein structures that shape caspase activity, specificity ...
    We present a comprehensive review of the caspases, their regulators and inhibitors from a structural and mechanistic point of view.Missing: paper | Show results with:paper
  5. [5]
    Identification and Characterization of a Novel Mammalian Caspase ...
    The catalytic domain consists of two subunits, p20 and p10, which, upon proteolytic processing of the proenzyme, assemble into a heterotetrameric complex, (p20/ ...
  6. [6]
    Targeting apoptotic caspases in cancer - ScienceDirect.com
    The active enzyme is thus a heterotetramer of the catalytic domains comprised of two large and two small subunits (p20/p10)2 (Fig. 2) [11]. Each caspase has a ...2. The Caspase Protease... · 7. Targeting Caspase... · 10. Caspase Independent Cell...
  7. [7]
    Identification and characterization of murine caspase-14, a new ...
    Like other caspases, caspase-14 has a conserved active site, pentapeptide QACRG. However, it lacks an NH2-terminal prodomain or a caspase recruitment domain, ...
  8. [8]
    A common allosteric site and mechanism in caspases - PMC
    We present a common allosteric mechanism for control of inflammatory and apoptotic caspases. Highly specific thiol-containing inhibitors of the human ...Missing: sheet QACRG
  9. [9]
    Structural basis for the activation of human procaspase-7 | PNAS
    These conformational changes are associated with the formation of an intact active-site cleft. Therefore, this mechanism represents a formerly unknown type ...
  10. [10]
    Caspase Functions in Cell Death and Disease - PMC
    Caspases are a family of endoproteases that provide critical links in cell regulatory networks controlling inflammation and cell death.
  11. [11]
    Old, new and emerging functions of caspases - PMC - NIH
    Caspases (cysteine-aspartic proteases) are proteolytic enzymes largely known for their role in controlling cell death and inflammation. The function of caspases ...Missing: paper | Show results with:paper
  12. [12]
    Caspases in cell death, inflammation and disease - PMC
    The caspases-1, 4, -5 and -11 are grouped as inflammatory caspases and share a CARD-domain at the N-terminal end. The apoptotic caspases can be further sub ...
  13. [13]
    Inflammasome activation and regulation: toward a better ... - Nature
    Jun 9, 2020 · Consequently, the active caspase-1 or caspase-8 leads to the maturation and secretion of inflammatory cytokines IL-1β and IL-18. The active ...
  14. [14]
    Recent advances in structural studies of NLRP3 and NLRP1 ...
    This review focuses on recent structural studies of NLRP3 and NLRP1, emphasizing the regulatory steps mediated by various activation and inhibitory factors.
  15. [15]
    Caspase activation: The induced-proximity model - PNAS
    Sep 28, 1999 · We review evidence for a hypothesis—the induced-proximity model—that describes how the first proteolytic signal is produced after adapter- ...
  16. [16]
    Interdimer processing mechanism of procaspase‐8 activation
    Here we show that dimerization of procaspase‐8 produces enzymatically competent precursors through the stable homophilic interaction of the procaspase‐8 ...Missing: seminal papers
  17. [17]
    Structural basis for dimerization of the death effector domain of the ...
    Nov 13, 2018 · We report two crystal structures of the dimeric DEDs of the F122A mutant of caspase-8, both of which illustrate a novel domain-swapped dimerization.Missing: seminal | Show results with:seminal
  18. [18]
    Human Caspases: Activation, Specificity, and Regulation - PMC - NIH
    Caspase activation is frequently followed by (auto)proteolytic cleavages called maturation events. Maturation is often an optional, chronologically distinct ...Human Caspases: Activation... · Initiator Caspases... · Executioner Caspases...<|control11|><|separator|>
  19. [19]
    Caspases: the proteases of the apoptotic pathway - PubMed - NIH
    Dec 24, 1998 · These death proteases are part of a proteolytic caspase cascade that is activated by diverse apoptotic stimuli from outside and inside of the cell.
  20. [20]
    X-Linked Inhibitor of Apoptosis Protein – A Critical Death Resistance ...
    A natural inhibitor of XIAP is SMAC/Diablo, which is released from mitochondria in apoptotic cells and displaces bound caspases from the BIR2/BIR3 domains of ...The Iap Family -- Structure... · Xiap-Induced Activation Of... · Xiap Overexpression In...<|control11|><|separator|>
  21. [21]
    Apoptosis-associated release of Smac/DIABLO from mitochondria ...
    Based largely upon in vitro studies, Smac/DIABLO appears to function by neutralizing the caspase-inhibitory properties of the IAP family of proteins, ...
  22. [22]
    Caspase-5: Structure, Pro-Inflammatory Activity and Evolution - PMC
    Apr 26, 2024 · The non-canonical inflammasome activators Caspase-4 and Caspase-5 are differentially regulated during immunosuppression-associated organ damage.
  23. [23]
    Catalytic activity and autoprocessing of murine caspase-11 mediate ...
    Jan 17, 2024 · These data reveal a previously undescribed role for Casp11 catalytic activity and autoprocessing in noncanonical inflammasome assembly.
  24. [24]
    Apoptosome: a platform for the activation of initiator caspases - Nature
    Sep 15, 2006 · In the structure, five distinct domains, CARD, three-layered α/β domain, helical domain I, winged-helix domain, and helical domain II, pack ...
  25. [25]
    Caspase-8 and Bid: Caught in the act between death receptors and ...
    This review deals with the events that enable cell death in type II cells, i.e., the signals that lead from death receptor stimulation to permeabilisation of ...
  26. [26]
    Caspase-8; regulating life and death - PMC - NIH
    This review describes the role of caspase-8 in the initiation of extrinsic apoptosis execution and the mechanism by which caspase-8 inhibits necroptosis.
  27. [27]
    The concept of intrinsic versus extrinsic apoptosis - Portland Press
    ... extrinsic apoptosis requires the proteolytic cleavage of BID ... Cleavage of BID by caspase 8 mediates the mitochondrial damage in the Fas pathway of apoptosis.
  28. [28]
    Regulation of the Apaf-1–caspase-9 apoptosome - PMC
    This review summarizes the accumulated structural and biochemical evidence that defines the origin of the first proteolytic signal of the intrinsic
  29. [29]
    The Apoptosome: Physiological, Developmental, and Pathological ...
    The intrinsic pathway involves the release of cytochrome c from the mitochondria into the cytosol. This leads to the formation of the apoptosome, the activation ...
  30. [30]
    Caspases and Their Substrates - PMC - NIH
    When activated, some caspases cleave hundreds, or perhaps thousands, of distinct target proteins. Such cleavage leads to all of the features that characterize ...
  31. [31]
    Many cuts to ruin: a comprehensive update of caspase substrates
    Nov 18, 2024 · The proteolytic cleavage can either lead to the functional inhibition or to the activation of these mediators. In some cases, it has been ...
  32. [32]
  33. [33]
    Caspase-dependent non-apoptotic processes in development - Nature
    May 19, 2017 · Caspase activation is locally regulated in subcellular compartments so that cellular remodeling during terminal differentiation is accomplished ...
  34. [34]
    The concealed side of caspases: beyond a killer of cells - PMC
    Dec 3, 2024 · Caspase-3 activation has been reported to induce apoptosis-like features during the differentiation of megakaryocytes, myoblasts, osteoblasts, ...
  35. [35]
    Physiological Functions of Caspases Beyond Cell Death
    These phenomena illustrate that activation of the caspase cascade does not necessarily lead to catastrophic proteolysis resulting in cell death, and each ...
  36. [36]
    Pyroptosis: mechanisms and diseases - Nature
    Mar 29, 2021 · Later, it was reported that caspase-1 or caspase-11/4/5 was activated during this process, and gasdermin D (GSDMD) was cleaved, and the N- ...Missing: seminal | Show results with:seminal
  37. [37]
    Pyroptosis in health and disease: mechanisms, regulation ... - Nature
    Sep 20, 2024 · In this review, we summarize the recent advancements in understanding the role of pyroptosis in disease, covering the related development ...
  38. [38]
    Mechanism of gasdermin D recognition by inflammatory caspases ...
    Jun 11, 2018 · Recent studies identified gasdermin D (GSDMD) as an effector for pyroptosis downstream of the inflammasome signaling pathways. Cleavage of GSDMD ...
  39. [39]
    Caspase-11 cleaves gasdermin D for non-canonical inflammasome ...
    Sep 16, 2015 · Here we show that gasdermin D is essential for caspase-11-dependent pyroptosis and interleukin-1β maturation.
  40. [40]
    Review The role of pyroptosis in metabolism and metabolic disease
    This paper reviews an in-depth study of the current advancements in understanding the role of pyroptosis in the progression of metabolic diseases.Missing: hyperactivation | Show results with:hyperactivation
  41. [41]
    Caspase-8 auto-cleavage regulates programmed cell death and ...
    Jan 21, 2022 · Caspase-8 is an initiator of death receptor-induced apoptosis and an inhibitor of RIPK3-MLKL-dependent necroptosis. In addition, caspase-8 ...
  42. [42]
    The double-edged functions of necroptosis | Cell Death & Disease
    Feb 27, 2023 · When caspase-8 is inhibited or absent, RIPK1 binds to RIPK3 via the RHIM domain, leading to RIPK3 phosphorylation and necrosome formation. An ...<|separator|>
  43. [43]
    Caspases as master regulators of programmed cell death: apoptosis ...
    Jun 24, 2025 · Caspases are crucial regulators of programmed cell death, mediating pathways such as apoptosis, pyroptosis, necroptosis and ferroptosis.
  44. [44]
    AG8 induces apoptosis and ferroptosis through regulating caspase ...
    Above all, AG8 induced apoptosis and ferroptosis through regulating caspase 3 and SLC7A11/GPX4 signaling pathway. Ferroptosis of LAR subtypes are easily to ...
  45. [45]
    Interplay of ferroptotic and apoptotic cell death and its modulation by ...
    Apr 29, 2025 · Ferroptotic death results from excessive lipid peroxidation, which manifests when the cellular antioxidant systems are overwhelmed in their ...
  46. [46]
    Ferroptosis, necroptosis, and pyroptosis in anticancer immunity
    Aug 10, 2020 · The induction of pyroptosis, ferroptosis, and necroptosis combined with ICIs showed synergistically enhanced antitumor activity, even in ICI-resistant tumors.
  47. [47]
    Insights on the crosstalk among different cell death mechanisms
    Feb 10, 2025 · This comprehensive review delves into the intricate molecular mechanisms, inducers, and inhibitors of the underlying prevalent forms of cell death.
  48. [48]
    Targeting regulated cell death: Apoptosis, necroptosis, pyroptosis ...
    Mar 19, 2025 · In this review, we first explore the mechanisms of apoptosis, necroptosis, pyroptosis, ferroptosis, and cuproptosis, along with the crosstalk ...
  49. [49]
    Ferroptosis in Cancer: Mechanism and Therapeutic Potential - PMC
    Apr 18, 2025 · By harnessing ferroptosis, cancer cells can be compelled to undergo lipid peroxidation-induced death, potentially improving therapeutic outcomes ...
  50. [50]
    Ferroptosis in the tumor microenvironment: mechanisms, advances ...
    Aug 21, 2025 · Inducing ferroptosis is considered an effective strategy to overcome tumor cell resistance to apoptosis. Notably, combining ferroptosis ...
  51. [51]
    The Inflammasome: A Caspase-1 Activation Platform Regulating ...
    Active caspase-1 is essential for the cleavage of pro-IL-1β and pro-IL-18 into their mature, biologically active forms. Mature IL-1β is implicated in multiple ...Missing: seminal papers
  52. [52]
    The role of inflammasomes in human diseases and their potential as ...
    Jan 5, 2024 · Inflammasomes are large protein complexes that play a major role in sensing inflammatory signals and triggering the innate immune response.<|separator|>
  53. [53]
    Regulation and function of the caspase-1 in an inflammatory ...
    Secreted IL-1α is not only the product of caspase-1 activity but is also involved in a positive feedback loop to activate NFκB in an autocrine or paracrine ...
  54. [54]
    Caspase-1/ASC Inflammasome-Mediated Activation of IL-1β–ROS ...
    We conclude that caspase-1/ASC inflammasomes play a significant role in the activation of IL-1β/ROS and NF-κB signaling of cytokine gene expression for T. cruzi ...
  55. [55]
    Inflammasomes and Signaling Pathways: Key Mechanisms in the ...
    Jun 19, 2025 · The non-canonical pathway bypasses this priming step and directly activates inflammatory caspases, leading to GSDMD cleavage, pore formation, ...
  56. [56]
    Caspase-1 activation, IL-1/IL-6 signature and IFNγ-induced ...
    During COVID-19, strong activation of the NLRP3-inflammasome and caspase-1 is induced with subsequent release of IL-1β and IL-18, although these cytokines ...Missing: overactivation sepsis
  57. [57]
    Activity-dependent synapse elimination requires caspase-3 activation
    Jun 10, 2025 · Our results reveal caspase-3 activation as a key step in activity-dependent synapse elimination during development and synapse loss in neurodegeneration.
  58. [58]
    Executioner Caspases and Their Diverse Non-apoptotic Roles
    Nov 4, 2015 · It is now certain that sublethal caspase activity plays a critical role in regulating neuronal pathfinding, pruning, and synaptic plasticity.
  59. [59]
    Hematopoietic stem cell responsiveness to exogenous signals ...
    Caspase-3 contributes to stem cell quiescence, dampening specific signaling events and thereby cell responsiveness to microenvironmental stimuli.Missing: asymmetric | Show results with:asymmetric
  60. [60]
    Role of the executioner caspases during lens development - PubMed
    Here, we test directly the role of executioner caspases (caspase-3, -6, and -7) in fiber cell differentiation. The distribution of mRNA, protein, and enzymatic ...Missing: myogenesis | Show results with:myogenesis<|control11|><|separator|>
  61. [61]
    Caspase 3 cleavage of Pax7 inhibits self-renewal of satellite cells
    Sep 8, 2015 · Here, we show activation of caspase 3 protease limits satellite cell self-renewal, through targeted cleavage and inactivation of Pax7 in a casein kinase (CK2)- ...Caspase 3 Cleavage Of Pax7... · Results And Discussion · Pax7 Protein Is Degraded Via...
  62. [62]
    Caspase activity mediates the differentiation of embryonic stem cells
    Here we report caspase-induced cleavage of Nanog in differentiating ES cells. Stem cells lacking the Casp3 gene showed marked defects in differentiation.Missing: 2025 | Show results with:2025
  63. [63]
    Non-genetic inactivation of caspase-3 and P53 increases cancer ...
    Oct 21, 2025 · These observations suggest that prolonged exposure to low doses of chemotherapeutic agents promotes the acquisition of multiple survival ...
  64. [64]
    Genetic control of programmed cell death in the nematode C. elegans
    The wild-type functions of the genes ced-3 and ced-4 are required for the initiation of programmed cell deaths in the nematode Caenorhabditis elegans.Missing: discovery | Show results with:discovery
  65. [65]
    A Cell That Dies during Wild-Type C. elegans Development Can ...
    Ellis and Horvitz (1986) identified a C. elegans gene, ced-3, that is necessary for almost all programmed cell deaths, so that in a ced-3 mutant, cells that ...
  66. [66]
    Molecular cloning of the interleukin-1 beta converting enzyme
    Interleukin-1 beta (IL-1 beta) mediates a wide range ... Molecular cloning of the interleukin-1 beta converting enzyme. Science. 1992 Apr 3;256(5053):97-100.Missing: Thornberry ICE
  67. [67]
    Induction of apoptosis in fibroblasts by IL-1β-converting enzyme, a ...
    We show here that overexpression of the murine ICE (mICE) gene or of the C. elegans ced-3 gene causes Rat-1 cells to undergo programmed cell death.
  68. [68]
    Study of apoptosis in vitro using the Xenopus egg extract ... - PubMed
    It was first shown by Newmeyer and colleagues in the 1990s that the molecular events of apoptosis could be reconstituted in vitro using Xenopus egg extracts ...Missing: discovery | Show results with:discovery
  69. [69]
    Structural basis for the oligomerization-facilitated NLRP3 activation
    Feb 7, 2024 · In this study, we determine the cryo-electron microscopy (cryo-EM) structures of human NLRP3 in the open oligomer, closed hexamer, and closed ...
  70. [70]
    Caspase family in autoimmune diseases - PubMed
    Regulating caspase activity holds the potential to restore immune balance in autoimmune diseases. Potential therapeutic approaches include small molecule ...
  71. [71]
    Are metacaspases caspases? - PMC - NIH
    We conclude that although metacaspases, paracaspases, and caspases contain a caspase fold and probably originated from a common ancestor gene, metacaspases and ...Missing: choanoflagellates | Show results with:choanoflagellates
  72. [72]
  73. [73]
    Identification of Novel Mammalian Caspases Reveals an Important ...
    Lineage-specific gene duplications were found for caspases-1, -3, and -12 in opossum and caspase-4 in primates. Other caspases were generally conserved in all ...
  74. [74]
    AtSERPIN1 is an inhibitor of the metacaspase AtMC1‐mediated cell ...
    Feb 3, 2017 · The hypersensitive response (HR) is a localized programmed cell death phenomenon that occurs in response to pathogen recognition at the site ...Summary · Introduction · Results · Discussion
  75. [75]
    Exploring the Roles of Metacaspases in Plants and Fungi - MDPI
    Dec 24, 2022 · Characteristics of fungal cell death include DNA fragmentation, cellular shrinkage, increased ROS production, and upregulation of metacaspase ...
  76. [76]
    Prokaryotic Caspase Homologs: Phylogenetic Patterns and ...
    Our data show a wide and scattered distribution of caspase homologs in prokaryotes with structurally and functionally diverse sub-groups.
  77. [77]
    Induction of apoptosis by double-stranded RNA was present in the ...
    Caspases in Metazoa: Phylogenetic analysis and domain organization. To broaden our understanding of the evolutionary history of caspases in Metazoans, we ...
  78. [78]
    The molecular cell death machinery in the simple cnidarian Hydra ...
    May 18, 2010 · Based on ESTs and the whole Hydra genome assembly, we have identified 15 caspases. We show that one is activated during apoptosis, four have ...
  79. [79]
    Intrinsic apoptosis is evolutionarily divergent among metazoans
    Non-bilaterian animals branch early, bilaterians are monophyletic and split across two main groups, the deuterostomian caspase-9 and the bilaterian caspase-2.
  80. [80]
    [PDF] Evolutionary dynamics of pro-inflammatory caspases in primates ...
    Aug 27, 2024 · Caspase-1 and related proteases are key players in inflammation and innate immunity. Here, we characterize the evolutionary history of ...
  81. [81]
    Evolutionary and Functional Analysis of Caspase-8 and ASC ...
    2024). Caspases (cysteine-dependent aspartyl-specific proteases; CASPs) are ancient regulators of programmed cell death, and some also drive inflammatory ...
  82. [82]
    Caspase Protocols in Mice - PMC - PubMed Central - NIH
    These methods include (1) determination of caspase activity using specific synthetic substrates designed to contain fluorogenic or chromogenic leaving groups ...
  83. [83]
    Z-VAD-FMK | Caspases - Tocris Bioscience
    Rating 5.0 (3) · $45 deliveryZ-VAD-FMK is a cell-permeable, irreversible pan-caspase inhibitor. Inhibits caspase processing and apoptosis induction in tumor cells in vitro (IC50 = 0.0015 - ...
  84. [84]
    A high throughput drug screen based on fluorescence resonance ...
    This FRET-based HTS method is a powerful tool for identifying anti-cancer compounds and can serve as a highly efficient platform for drug discovery.Missing: luminescence | Show results with:luminescence
  85. [85]
    Detection of localized caspase activity in early apoptotic cells by ...
    Jan 17, 2002 · One recently described method is the use of the cell-permeable fluorogenic caspase 3/7 substrate PhiPhiLux (15). This reagent consists of an 18- ...
  86. [86]
    The caspase 3 sensor Phiphilux G2D2 is activated non-specifically ...
    Phiphilux is a new generation cell permeable probe which emits fluorescence in the red (G2D2) or green (G1D2) when cleaved by active caspase 3. Its specificity ...
  87. [87]
    An Improved Cell-Penetrating, Caspase-Activatable, Near-Infrared ...
    Several imaging probes have been developed that detect caspase activity, including examples of imaging probes that incorporate fluorescence resonance energy ...
  88. [88]
    An Improved Cell-Penetrating, Caspase-Activatable, Near-Infrared ...
    KcapQ represents an improved effector caspase-activatable NIF probe for enhanced non-invasive analysis of apoptosis in whole cells and live animals.
  89. [89]
    Spatio-temporal activation of caspase revealed by indicator that is ...
    Indicator molecules for caspase-3 activation have been reported that use fluorescence resonance energy transfer (FRET) between an enhanced cyan fluorescent ...
  90. [90]
    A FRET biosensor for necroptosis uncovers two different modes of ...
    Oct 26, 2018 · We previously developed a FRET biosensor termed SCAT (a Sensor for Caspase Activation based on FRET). SCAT1 and SCAT3 comprise peptides ...
  91. [91]
    Bioluminescence imaging of caspase-3 activity in mouse liver
    In the present work, we established a stable mouse model by hydrodynamics methods to study the activity of caspase-3 and evaluate the effect of the apoptosis ...Missing: PET tracers
  92. [92]
    Positron emission tomography imaging of drug-induced tumor ...
    In summary, we have characterized [18F]ICMT-11 as a caspase-3/7 specific PET imaging radiotracer for the assessment of tumor apoptosis that could find utility ...
  93. [93]
    Development of caspase-3-selective activity-based probes for PET ...
    Aug 8, 2024 · In this study we developed novel 18F-labelled PET tracers for the imaging of caspase-3 activation for treatment response evaluation.
  94. [94]
    Large transient assemblies of Apaf1 constitute the apoptosome in cells
    Oct 24, 2025 · Cryo-EM structure of caspase-8 Tandem DED filament reveals assembly and regulation mechanisms of the death-inducing signaling complex. Mol ...
  95. [95]
    Mitochondrial dynamics and pore formation in regulated cell death ...
    Oct 1, 2025 · We highlight the impact of advanced single-molecule and correlative microscopy techniques which have driven recent progress in understanding the ...Missing: apoptosomes | Show results with:apoptosomes