Fact-checked by Grok 2 weeks ago

Pyroptosis

Pyroptosis is a form of that is inflammatory and lytic in nature, triggered by the detection of pathogens or endogenous danger signals within the , leading to permeabilization, swelling, rupture, and the release of pro-inflammatory cytokines such as interleukin-1β (IL-1β) and IL-18. This process is mediated primarily by gasdermin family proteins, particularly gasdermin D (GSDMD), which, upon cleavage by inflammatory (e.g., caspase-1, -4, -5, or -11), oligomerize to form pores in the plasma membrane, disrupting ion balance and facilitating the extrusion of danger-associated molecular patterns (DAMPs) and alarmins. Unlike , which is immunologically silent, pyroptosis amplifies innate immune responses but can contribute to tissue damage if uncontrolled. The mechanisms of pyroptosis involve two main pathways: the canonical pathway, where sensor proteins like NLRP3 form in response to stimuli such as ATP or bacterial toxins, activating caspase-1 to process GSDMD and pro-IL-1β; and the non-canonical pathway, activated directly by (LPS) from via caspase-4/5 (in humans) or caspase-11 (in mice). Additional executors include other gasdermins like GSDME, which can be cleaved by caspase-3 during to induce pyroptotic features, and ninjurin-1 (NINJ1), which aids in the final membrane rupture. These pores, approximately 10–20 nm in diameter, allow the passive release of cellular contents while maintaining initial membrane integrity for secretion. First described in the early as a distinct form of death induced by bacterial pathogens like and , pyroptosis was formally named in 2001 to highlight its fever-inducing inflammatory properties, derived from the Greek words "pyro" (fire) and "ptosis" (falling). The identification of gasdermins as the central effectors in 2015 marked a breakthrough, linking pyroptosis to broader gasdermin-mediated pyroptotic pathways. In host defense, pyroptosis eliminates intracellular pathogens and signals danger to neighboring cells, enhancing immunity against infections like or . However, excessive pyroptosis drives pathology in conditions such as , where it exacerbates storms; like cryopyrin-associated periodic syndromes (CAPS) and (FMF); and chronic disorders including , neurodegeneration, and cancer, where it may suppress tumors by boosting immunity or promote progression via . Therapeutic strategies targeting pyroptosis, such as GSDMD inhibitors, are under investigation to balance its protective and detrimental effects.

Overview

Definition and Key Features

Pyroptosis is a lytic and pro-inflammatory form of that is dependent on and mediated by gasdermin proteins, resulting in plasma membrane rupture and the release of intracellular contents without the formation of apoptotic bodies. Unlike non-lytic pathways, pyroptosis leads to the extrusion of pro-inflammatory mediators such as interleukin-1β (IL-1β) and IL-18, which amplify immune responses. The term "pyroptosis" was coined in 2001 by Cookson and Brennan to describe a caspase-1-dependent observed in macrophages infected with , derived from the Greek words "" (fire or fever) and "ptosis" (falling), emphasizing its fever-inducing inflammatory nature. Key features of pyroptosis include rapid cell swelling due to osmotic imbalance, formation of pores in the plasma membrane by the N-terminal fragments of gasdermins—primarily gasdermin D (GSDMD)—and subsequent cell lysis that generates pyroptotic bodies. GSDMD, a member of the gasdermin family, is cleaved by inflammatory (such as caspase-1, -4, -5, or -11) to release its pore-forming N-terminal domain, which oligomerizes to create ~20 nm diameter pores that permeabilize the membrane. This process is typically triggered by activation in response to microbial or endogenous danger signals, distinguishing pyroptosis as an innate immune defense mechanism. The primary outcomes of pyroptosis involve the release of damage-associated molecular patterns (DAMPs), such as high-mobility group box 1 (), alongside cytokines, which propagate and recruit immune cells to sites or damaged tissues. This contrasts with non-lytic forms of by actively promoting a hyperinflammatory state rather than silent clearance, thereby enhancing host defense but potentially contributing to if dysregulated.

Distinction from Other Cell Death Pathways

Pyroptosis differs fundamentally from apoptosis in its pro-inflammatory nature and lytic execution. While apoptosis is an anti-inflammatory process characterized by non-lytic cell shrinkage, chromatin condensation, membrane blebbing, and formation of apoptotic bodies that prevent content leakage, pyroptosis involves rapid cell swelling, membrane rupture via gasdermin D (GSDMD) pores, and release of intracellular contents, including pro-inflammatory cytokines like IL-1β and IL-18. Apoptosis relies on caspase-3 and caspase-7 activation without GSDMD involvement or inflammasome priming, serving to maintain tissue homeostasis without alerting the immune system, whereas pyroptosis lacks caspase-3/7 activation and is driven by caspase-1 or caspase-11 to amplify innate immune responses. In contrast to necroptosis, another lytic form of , pyroptosis is specifically dependent on activation and caspases-1/11, leading to targeted release and GSDMD-mediated pore formation. Necroptosis, triggered by death receptor signaling such as TNF-α, involves the /RIPK3/MLKL pathway, where phosphorylated MLKL oligomerizes to disrupt the plasma membrane, but it does not require inflammasome priming or result in IL-1 family maturation. Both pathways cause membrane permeabilization and pro-inflammatory (DAMP) release, yet pyroptosis's reliance on pathogen-associated molecular patterns (PAMPs) or DAMPs distinguishes it as a primary defense against microbial , while necroptosis acts as a mechanism when is inhibited. Pyroptosis also contrasts with , which features iron-dependent as its core mechanism without significant involvement of or release. Ferroptosis leads to lytic through accumulation of lipid hydroperoxides and depletion, often resulting in mitochondrial shrinkage but eventual membrane rupture, and it engages pro-inflammatory pathways through DAMP exposure like HMGB1. In pyroptosis, plays a negligible role, with execution centered on GSDMD pores and caspase-dependent processing, emphasizing its role in innate immunity rather than ferroptosis's association with responses in conditions like ischemia or cancer. The following table summarizes key distinctions across these pathways:
AspectPyroptosisApoptosisNecroptosisFerroptosis
TriggersPAMPs/DAMPs via inflammasomesDeath ligands (e.g., TNF), DNA damageTNF-α, viral infection, TLRsIron overload, lipid peroxidation inducers
ExecutionersGSDMD pores (caspase-1/11-cleaved)Caspases-3/7/9MLKL (RIPK3-phosphorylated)Lipid peroxidation (no specific protein)
MorphologyLytic: swelling, membrane ruptureNon-lytic: shrinkage, apoptotic bodiesLytic: swelling, membrane ruptureLytic: mitochondrial shrinkage, membrane rupture
Inflammatory OutcomePro-inflammatory (IL-1β/IL-18 release)Anti-inflammatory (no content leakage)Pro-inflammatory (DAMP release)Pro-inflammatory (HMGB1/ATP)
Physiological RoleInnate immunity, pathogen clearanceTissue homeostasis, developmentBackup death, inflammationTumor suppression, oxidative defense

History

Initial Discovery

The initial observation of what would later be recognized as pyroptosis occurred in 1992, when Zychlinsky and colleagues reported that infection of macrophages with the Gram-negative bacterium Shigella flexneri triggered a form of programmed cell death resembling apoptosis, characterized by DNA fragmentation and cell lysis. This death was distinct from bacterial replication in non-phagocytic cells and was proposed to contribute to the inflammatory response in shigellosis by limiting intracellular pathogen growth. Subsequent work in 1994 by the same group demonstrated that Shigella-infected macrophages released mature interleukin-1β (IL-1β), linking the cell death process to pro-inflammatory cytokine production during infection. Parallel early observations in the 1990s included death induced by Bacillus anthracis lethal toxin, which was later shown to activate caspase-1 and promote inflammatory cell lysis similar to Shigella-induced death. Further characterization in 1997 revealed that this death was dependent on the interleukin-1β-converting enzyme (ICE), now known as caspase-1, as inhibition of ICE with the peptide YVAD-CHO blocked both and IL-1β processing in Shigella-infected murine macrophages. This finding highlighted caspase-1's role in the pathway but also contributed to initial confusion with classical , given the shared involvement of and morphological similarities like nuclear condensation. Experiments using caspase-1 knockout mice, reported in 1998, confirmed the enzyme's necessity, as macrophages from these animals resisted Shigella-induced death while remaining susceptible to other apoptotic stimuli. In 2000, Brennan and Cookson extended these observations to Salmonella infections, showing that the bacterium induced rapid, caspase-1-dependent macrophage death accompanied by IL-1β secretion but lacking typical apoptotic features such as caspase-3 activation or phosphatidylserine exposure. This process limited Salmonella replication by promoting bacterial expulsion from dying cells, providing early evidence of its protective role in host defense. The following year, Cookson and Brennan coined the term "pyroptosis" to describe this distinct, pro-inflammatory form of cell death, emphasizing its rapid kinetics, inflammatory cytokine release, and divergence from non-inflammatory apoptosis. Key validation came from studies in caspase-1-deficient mice, which exhibited markedly reduced acute and impaired clearance of following oral infection, underscoring pyroptosis's contribution to orchestrating immune responses against intracellular bacteria.

Major Milestones and Recent Advances

The identification of gasdermin D (GSDMD) as the key executor of pyroptosis marked a pivotal breakthrough in 2015, when independent studies by the Shi, Kayagaki, and Ding groups demonstrated that GSDMD is cleaved by inflammatory (caspase-1 in the canonical pathway and caspase-11/4/5 in the non-canonical pathway) to release its N-terminal domain, which forms plasma membrane pores leading to lysis and interleukin-1β release. In 2016, the Ding group further elucidated the pore-forming mechanism of the GSDMD N-terminal fragment, confirming its oligomerization into 10- to 20-subunit pores approximately 16-24 nm in diameter, which disrupts osmotic balance and triggers pyroptotic . Building on this, the non-canonical inflammasome pathway was more precisely characterized in 2014, revealing that mouse caspase-11 (human orthologs caspase-4/5) directly senses cytosolic lipopolysaccharide from Gram-negative bacteria and cleaves GSDMD to induce pyroptosis independent of canonical inflammasome sensors like NLRP3. This pathway highlighted pyroptosis's role beyond inflammasome activation, emphasizing direct pathogen detection as a rapid innate immune response. Research in 2017 expanded pyroptosis beyond GSDMD dependence, with studies showing that gasdermin E (GSDME) mediates an alternative form of pyroptosis in cancer cells through caspase-3 cleavage, particularly in response to chemotherapeutic agents, thereby enhancing antitumor immunity by releasing damage-associated molecular patterns. A 2024 study uncovered pyroptosis's physiological role in neural development, where AIM2 inflammasome-driven GSDMD activation clears DNA-damaged cells during cortical , preventing neurodevelopmental disorders like anxiety-like behaviors. As of 2025, key advances include the discovery that S-palmitoylation of at multiple residues, catalyzed by ZDHHC enzymes, sequentially regulates assembly and activation, offering new targets for modulating pyroptotic in diseases like atherosclerosis. Plant-derived compounds such as and have emerged as selective pyroptosis inducers in drug-resistant tumors, activating GSDME-mediated death while sparing healthy cells and synergizing with . Additionally, small-molecule GSDMD inhibitors like AI-screened pore blockers (e.g., SK56) and repurposed drugs targeting N-terminal oligomerization have advanced to preclinical trials, demonstrating efficacy in attenuating excessive pyroptosis in and autoinflammatory conditions without compromising clearance. Overall, these milestones reflect a in pyroptosis research from its initial focus on to its broader implications in cancer therapy, neurodevelopment, and chronic inflammation, with therapeutic strategies increasingly targeting gasdermin pores for precision modulation.

Cellular Characteristics

Morphological Changes

Pyroptosis begins with the formation of pores in the plasma membrane mediated by gasdermin D (GSDMD), which initiates a cascade of structural alterations in the affected cell. In the early phase, these pores, measuring 10-20 nm in diameter, permit non-selective ion fluxes, including initial potassium (K⁺) efflux followed by sodium (Na⁺) and chloride (Cl⁻) influx, leading to osmotic imbalance and rapid cell swelling. This swelling is observable through live-cell microscopy and can result in up to a 1.5-fold increase in cell volume prior to further progression. As pyroptosis advances, the accumulating osmotic pressure causes the plasma membrane to rupture, releasing intracellular contents such as lactate dehydrogenase (LDH), which serves as a quantifiable marker of membrane integrity loss. Electron microscopy visualizations confirm the presence of these GSDMD-induced pores on the membrane surface, contributing to the lytic nature of the process. In the late phase, cells form balloon-like protrusions or pyroptotic bodies, characterized by membrane blebbing and eventual fragmentation into corpse-like structures, distinct from the apoptotic shrinkage. Nuclear changes during pyroptosis include DNA condensation without the internucleosomal fragmentation typical of apoptosis, as observed via live-cell imaging techniques that track these morphological dynamics in real time. These alterations culminate in the complete lysis of the cell, facilitating the release of proinflammatory mediators while preserving key subcellular components within the resulting pyroptotic remnants.

Biochemical and Molecular Markers

Pyroptosis is characterized by specific biochemical and molecular signatures that distinguish it from other forms of , primarily involving the activation of inflammatory and the pore-forming protein gasdermin D (GSDMD). The hallmark marker is the cleavage of GSDMD into its N-terminal fragment (GSDMD-NT), which oligomerizes to form plasma membrane pores, leading to cell lysis and release of intracellular contents. This cleavage occurs at aspartate residue 275 in humans (D275) or 276 in mice (D276) by active caspase-1, caspase-11 (in mice), or caspase-4/5 (in humans), producing the ~30 kDa GSDMD-NT fragment detectable by analysis of cell lysates or supernatants. Another key indicator is the processing of pro-caspase-1 into its active p20 subunit (along with p10), which drives GSDMD cleavage and is similarly identified via , confirming activation. Additionally, the maturation and extracellular release of pro-inflammatory cytokines interleukin-1β (IL-1β) and IL-18, processed by caspase-1, serve as soluble markers enriched in culture supernatants or serum, reflecting the lytic nature of pyroptosis. Detection of these markers relies on established assays tailored to pyroptotic events. Western blotting remains the gold standard for visualizing cleaved GSDMD-NT and the p20 subunit of caspase-1/11, often using antibodies specific to the N-terminal domain or neo-epitopes generated post-cleavage, allowing quantification of pyroptosis induction in response to stimuli like lipopolysaccharide (LPS). Cytokine release is quantified via enzyme-linked immunosorbent assay (ELISA) for mature IL-1β and IL-18 in supernatants, providing a sensitive measure of inflammasome-driven pyroptosis without requiring cell lysis. Flow cytometry distinguishes pyroptotic cells by their Annexin V-negative/propidium iodide (PI)-positive profile, indicating membrane permeabilization and loss of integrity prior to full lysis, in contrast to apoptotic cells which are Annexin V-positive/PI-negative; this method can be combined with antibodies against cleaved GSDMD for enhanced specificity. In vivo, pyroptosis manifests through systemic indicators of cell damage and inflammasome activity. Elevated serum lactate dehydrogenase (LDH) levels, released due to GSDMD pore-mediated membrane rupture, serve as a non-specific but quantitative proxy for pyroptotic in tissues or circulation, measurable by enzymatic assays in animal models of or sterile . ASC specks—oligomeric complexes of apoptosis-associated speck-like protein containing a (ASC)—are released extracellularly following pyroptosis and can be detected in tissues via using anti-ASC antibodies, highlighting persistent inflammasome foci that amplify . To differentiate pyroptosis from or other necrotic pathways, functional validation assays are essential. Genetic of GSDMD abolishes pore formation and IL-1β release while preserving caspase-1 and processing, confirming GSDMD dependency and ruling out alternative death modes. Pharmacological inhibitors like disulfiram, which covalently modifies GSDMD at 191/192 to block NT oligomerization, selectively prevent pyroptotic and LDH release without interfering with apoptotic caspase-3 , enabling mechanistic in experimental settings. As of 2025, advances in understanding regulation include its S-palmitoylation at residues (e.g., Cys126) by ZDHHC enzymes such as ZDHHC7, which promotes assembly and serves as an early marker of pyroptotic priming; this modification is detected using indirect methods like acyl-biotin exchange (ABE) assays in immunoblotting and .

Molecular Mechanisms

Canonical Inflammasome Pathway

The canonical inflammasome pathway is the primary mechanism by which pyroptosis is induced in response to microbial and sterile stimuli, involving the assembly of pattern recognition receptors such as NLRP3 or AIM2 with the adaptor protein ASC and pro-caspase-1 to form an active inflammasome complex. This pathway culminates in the activation of caspase-1, which processes pro-inflammatory cytokines and executes cell lysis through gasdermin D (GSDMD) pore formation. Unlike other cell death modalities, this process is tightly regulated to balance innate immune defense with tissue integrity. The pathway requires a two-step process for . In the priming phase (signal 1), pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs) are recognized by Toll-like receptors (TLRs), leading to translocation and transcriptional upregulation of , pro-IL-1β, and pro-IL-18. This step prepares the cell by increasing the availability of components without immediate . The phase (signal 2) is triggered by diverse stimuli, including potassium (K⁺) efflux, lysosomal membrane destabilization, or mitochondrial (mtROS) production, which promote oligomerization and recruitment of ASC via PYRIN domains. For AIM2, double-stranded DNA from pathogens or damaged cells directly binds and activates the sensor, bypassing some priming requirements but often integrating with signals. Inflammasome assembly results in the recruitment and autocleavage of pro-caspase-1 into its active form, a p20/p10 heterotetramer, which then cleaves substrates. This active caspase-1 processes pro-IL-1β and pro-IL-18 into their mature, secreted forms, amplifying inflammation. Critically, caspase-1 also cleaves GSDMD at Asp275 (in humans) or Asp276 (in mice), separating the N-terminal fragment (GSDMD-NT, residues 1-275/276) from the inhibitory C-terminal domain (GSDMD-CT). The GSDMD-NT domain oligomerizes and inserts into the plasma membrane, forming non-selective pores approximately 10-20 nm in diameter that permit the efflux of ions, water influx, and cytokine release, ultimately causing osmotic cell swelling and lysis characteristic of pyroptosis. This pore formation is essential for pyroptotic execution, as GSDMD-deficient cells resist lysis despite inflammasome activation. Representative triggers illustrate the pathway's versatility. The bacterial toxin nigericin induces K⁺ efflux from cells, a key signal for NLRP3 activation in primed macrophages. Similarly, monosodium urate (MSU) crystals, associated with , destabilize lysosomes to release , promoting NLRP3 assembly and downstream pyroptosis. These examples highlight how environmental and microbial cues converge on common intracellular signals to drive the canonical response.

Non-Canonical Inflammasome Pathway

The non-canonical inflammasome pathway represents a distinct of pyroptosis activation in response to intracellular (LPS) from , primarily mediated by caspase-11 in mice and caspases-4 and -5 in humans. Unlike the canonical pathway, this route operates independently of inflammasome sensors such as NLRP3 or NLRC4 and does not require the adaptor protein ASC or prior transcriptional priming signals. It enables rapid detection of cytosolic LPS, triggering inflammatory activation and subsequent cell lysis to restrict replication. The activation process begins with direct binding of LPS to the catalytic p20 subunit of pro-caspase-11/4/5 in the , facilitated by an exosite on the that recognizes the moiety of LPS. This interaction induces oligomerization of the into dimers or higher-order structures, promoting autocleavage at specific aspartate residues (e.g., Asp289 in ) to generate the active heterotetramer. Autocleavage is essential for activity but occurs without the need for external processing, distinguishing this pathway as a single-step response to cytosolic bacterial components. The process is particularly prominent during Gram-negative bacterial infections where LPS is released into the host , such as upon . Upon activation, caspase-11/4/5 directly cleaves gasdermin D (GSDMD) at Asp276 () or Asp275 (), liberating the N-terminal GSDMD fragment that oligomerizes to form membrane pores. These pores cause osmotic cell swelling, membrane rupture, and pyroptotic , releasing intracellular contents including alarmins. Indirectly, GSDMD pore formation leads to efflux, which can secondarily activate the canonical NLRP3 and caspase-1, enabling maturation and secretion of interleukin-1β (IL-1β); however, IL-1β release is limited compared to canonical activation. Key differences from the canonical pathway include the absence of efflux as a primary trigger, reliance on direct LPS sensing rather than two-step , and a stronger emphasis on pyroptosis over processing in Gram-negative contexts. Experimental evidence for this pathway emerged from studies in the early demonstrating that caspase-11-deficient mice are highly resistant to lethal endotoxin induced by high-dose LPS, even in the absence of (TLR4) signaling, which handles extracellular LPS detection. of synthetic LPS into macrophages from wild-type but not caspase-11 knockout cells induced pyroptosis, confirming direct cytosolic sensing. Similarly, human monocytes and macrophages respond to cytosolic LPS via caspases-4 and -5, with knockdown protecting against pyroptotic responses to like . These findings established caspase-11/4/5 as critical sensors for intracellular bacterial threats. Recent advances highlight cross-talk with guanylate-binding proteins (GBPs), interferon-inducible effectors that target pathogen-containing vacuoles and facilitate LPS exposure to caspases-11/4/5. GBPs, such as GBP1 and GBP2, disrupt bacterial outer membranes or endosomal compartments, releasing LPS into the to enhance non-canonical activation during infections like or . This GBP-mediated targeting amplifies host defense by promoting pyroptosis specifically against cytosolic invaders, with disruptions in GBP function impairing caspase-11 responses . Studies as recent as 2023 underscore GBPs' role in integrating non-canonical signaling with broader innate immunity, including modulation, to fine-tune inflammatory outcomes.

Caspase-3-Dependent Pathway

The caspase-3-dependent pathway of pyroptosis represents a non-canonical mechanism that intersects with , where executioner caspase-3 cleaves gasdermin E (GSDME) to trigger lytic cell death instead of the typical apoptotic fragmentation. This pathway was first identified in 2017 by Wang et al., who showed that in gastric s expressing high levels of GSDME, chemotherapeutic agents like activate the apoptotic cascade, leading caspase-3 to cleave GSDME and induce pyroptotic features such as membrane rupture and release. Independently, Zhang et al. in 2018 demonstrated that cleaved GSDME functions as the key effector downstream of caspase-3 in various lines treated with , confirming the switch from to pyroptosis in GSDME-expressing cells. Mechanistically, stimuli such as chemotherapeutic drugs or other inducers (e.g., ) engage the extrinsic pathway via or the intrinsic pathway via , culminating in caspase-3 activation from its pro-form. Active caspase-3 then specifically cleaves GSDME at the Asp270 residue, liberating the N-terminal domain (GSDME-N) that assembles into oligomeric pores on the plasma membrane, disrupting osmotic balance and causing cell swelling and . This process can be represented as: \text{pro-caspase-3} \to \text{active caspase-3} \to \text{GSDME-NT} \to \text{membrane pores} Unlike GSDMD-mediated pyroptosis in the canonical inflammasome pathway, GSDME cleavage by caspase-3 does not directly mature IL-1β or IL-18, but the pore formation enables passive release of these cytokines along with other intracellular contents. Cell-type specificity is a hallmark of this pathway, as it predominates in tissues or cells with elevated GSDME expression, such as gastric and breast cancer cells, where it converts intended apoptotic responses into pyroptotic ones, enhancing antitumor immunity through inflammation. In neurons, which also express high GSDME, this mechanism contributes to pyroptotic cell death under stress conditions, shifting from quiet apoptosis to immunogenic lysis. In contrast, many tumor cells with low or silenced GSDME undergo standard apoptosis without pore formation, underscoring GSDME's role as a molecular switch. The outcomes of caspase-3/GSDME-mediated pyroptosis include rapid cell lysis followed by secondary necrosis-like , driven by the efflux of damage-associated molecular patterns (DAMPs) and alarmins that recruit immune cells and amplify local responses. While direct IL-1β processing is absent, indirect release occurs through cellular damage signals, promoting broader inflammatory cascades without the need for assembly. Recent research has extended this pathway's relevance to neurodegeneration, showing that in HIV-infected brains, convergent caspase-1 and -3 activation cleaves GSDME to induce neuronal pyroptosis, contributing to cognitive decline in HIV-associated neurocognitive disorders.

Emerging Gasdermin-Centric Pathways

Recent discoveries have revealed gasdermin-centric pathways in pyroptosis that operate independently of the classical caspase-1/11 or caspase-3 activations, highlighting diverse proteolytic and post-translational mechanisms that fine-tune inflammatory . These emerging routes often involve alternative proteases or integrated programs, expanding the scope of pyroptosis beyond canonical to include immune cell interactions and stress responses. Such pathways underscore the adaptability of gasdermins in host defense and , with implications for targeted therapies. One prominent GSDMD-independent mechanism involves cleavage of gasdermin B (GSDMB) by granzyme A secreted from natural killer (NK) cells and cytotoxic T lymphocytes during immune synapse formation. Granzyme A proteolytically activates GSDMB by cleaving at specific lysine residues (e.g., K229 or K244), releasing the N-terminal fragment that oligomerizes to form plasma membrane pores, thereby inducing pyroptosis in target cells such as tumor cells. This process enhances anti-tumor immunity by promoting the release of inflammatory contents from lysed cells, as demonstrated in esophageal carcinoma models where GSDMB expression correlates with NK cell-mediated cytotoxicity. Similarly, granzyme B can cleave GSDMB at distinct sites, further diversifying pyroptotic execution in immune responses against infected or malignant cells. In neutrophils, GSDMD activation occurs independently of caspases through cleavage by neutrophil elastase released during lysosomal permeabilization in aging or activated cells. This targets GSDMD at non-canonical sites, facilitating pore formation and pyroptosis that contributes to inflammatory or exacerbation in conditions like . Such elastase-mediated processing allows neutrophils to undergo lytic death without inflammasome involvement, releasing contents while amplifying local at immune synapses with pathogens or other immune cells. A key integration of pyroptosis with other cell death modalities occurs in PANoptosis, where ZBP1 senses or sterile triggers to assemble a PANoptosome complex incorporating RIPK3 and , concurrently driving pyroptosis, , and necroptosis. In infection, ZBP1 recruits RIPK3 to phosphorylate MLKL for necroptosis while enabling GSDMD cleavage for pyroptotic pores, amplifying inflammatory signaling without exclusive reliance on . This unified pathway, observed in diverse infections, ensures robust host defense by lytic but can exacerbate tissue damage if dysregulated. Post-translational modifications have emerged as critical regulators of gasdermin function, with recent studies () elucidating their roles in pore stability and inhibition. Palmitoylation of the GSDMD N-terminal fragment (GSDMD-NT) at conserved residues (e.g., Cys191/192) promotes its membrane translocation and oligomerization, stabilizing pores essential for pyroptosis execution in response to or . This lipid modification, enhanced by , is indispensable for GSDMD's insertion into lipid bilayers, as mutants lacking palmitoylation fail to induce cell lysis. Conversely, ubiquitination of GSDMD-NT modulates its activity; for example, K48-linked ubiquitination by E3 ligases like TRIM21 can inhibit oligomerization by altering membrane targeting, thereby suppressing excessive pyroptosis in inflammatory settings. These modifications provide dynamic control points, with 2025 research highlighting ubiquitination's role in directing GSDMD degradation or relocation to prevent uncontrolled . Beyond GSDMD, GSDMB in gastrointestinal epithelia undergoes activation via caspase-3 or cleavage during apoptosis-pyroptosis crosstalk, or by bacterial proteases like those from , leading to epithelial pyroptosis and barrier disruption. In the gut, processes GSDMB at apoptotic sites to release its pore-forming domain, exacerbating in conditions like , while bacterial enzymes exploit this for pathogen escape. This dual activation underscores GSDMB's context-specific role in mucosal immunity. Alternative cleavage sites on GSDMD itself, such as those targeted by in (TNF)-induced death, represent another emerging pathway. cleaves GSDMD at the canonical Asp275 (humans)/Asp276 (mice) site, generating the pore-forming N-terminal fragment and promoting lethality in TNF-challenged cells, independent of caspase-1. This integrates extrinsic apoptosis signals with pyroptosis, as evidenced in genetic models where caspase-8/GSDMD double deficiency protects against TNF shock, highlighting its relevance in .

Regulation

Activators and Triggers

Pyroptosis is triggered by a diverse array of upstream signals that alert the to microbial invasion, cellular damage, or environmental perturbations, leading to the activation of or related pathways. These activators encompass pathogen-associated molecular patterns (PAMPs), damage-associated molecular patterns (DAMPs), and synthetic or environmental stimuli, which converge on sensors such as , AIM2, NLRC4, or non-canonical to initiate the process. Microbial triggers primarily involve PAMPs recognized by cytosolic sensors. Lipopolysaccharide (LPS) from directly binds and activates human caspase-4/5 or murine caspase-11 in the non-canonical pathway, independent of TLR4 priming, thereby sensing cytosolic bacterial presence. , a component of bacterial flagella, is detected by neuronal apoptosis inhibitory proteins (NAIPs), which recruit and activate NLRC4 to form an complex, as demonstrated in responses to infection. Similarly, double-stranded DNA (dsDNA) or from viruses and engages the AIM2 sensor, which oligomerizes upon binding to initiate caspase-1 activation, a mechanism pivotal in defending against intracellular pathogens like . Endogenous activators arise from host-derived danger signals released during cellular stress or injury. (mtDNA), extruded into the following mitochondrial damage, acts as a DAMP to stimulate the , contributing to sterile inflammation in conditions like non-alcoholic . Extracellular ATP, often liberated from dying cells, binds the P2X7 receptor to induce potassium efflux and NLRP3 activation, amplifying inflammatory responses. Cholesterol crystals, prevalent in atherosclerotic plaques, are phagocytosed by macrophages, causing lysosomal destabilization and engagement, which drives IL-1β secretion and promotes atherogenesis. In sterile inflammatory diseases such as , monosodium urate crystals similarly serve as DAMPs to trigger NLRP3-dependent pyroptosis. Environmental and pharmacological factors can mimic pathogenic signals to provoke pyroptosis. Nigericin, a potassium derived from , depletes intracellular K⁺, serving as a canonical second signal for activation in primed macrophages, widely used to model bacterial toxin effects. Lysosomal destabilizers, such as L-leucyl-L-leucine methyl ester (LLOMe), rupture lysosomes to release cathepsins, indirectly activating and inducing pyroptosis in immune cells. Cell-specific mechanisms fine-tune trigger responsiveness. For NLRP3, priming via NF-κB signaling—often induced by Toll-like receptor ligands like LPS—upregulates NLRP3 and pro-IL-1β expression, sensitizing cells to subsequent activators such as ATP or nigericin. Guanylate-binding proteins (GBPs), interferon-inducible effectors, facilitate LPS delivery to caspase-11 by targeting bacterial outer membranes and promoting lipopolysaccharide aggregation, essential for non-canonical inflammasome sensing of Gram-negative invaders. Recent advances highlight viral contributions to pyroptosis triggers. infection induces ZBP1-dependent PANoptosis in bystander cells via secreted inflammatory factors such as 2′3′-cGAMP, TNF-α, and IFN-β, exacerbating pathology, as reported in 2025 studies.

Inhibitors and Modulatory Mechanisms

Pyroptosis is tightly regulated by endogenous inhibitors that prevent unwarranted of its components. The C-terminal (GSDMD-CT) of gasdermin D (GSDMD) exerts autoinhibition by to and masking the pore-forming N-terminal (GSDMD-NT), thereby suppressing oligomerization and membrane perforation until proteolytic cleavage occurs. Similarly, post-translational modifications of , such as at Ser803 within its leucine-rich repeat , disrupt the interaction with NEK7, a essential for NLRP3 oligomerization and inflammasome assembly, thus inhibiting downstream caspase-1 and pyroptosis. Ubiquitination of by E3 ligases like MARCH5 on mitochondrial surfaces promotes NLRP3-NEK7 complex formation under basal conditions but can be reversed by deubiquitinases to dissociate NEK7 and attenuate signaling. Pharmacological agents target key steps in pyroptosis to modulate its execution. Disulfiram, an FDA-approved , covalently modifies Cys191 (human) or Cys192 () in GSDMD-NT, blocking formation and thereby preventing IL-1β release and without affecting upstream activation. Necrosulfonamide (NSA), a small-molecule , similarly targets a reactive in GSDMD to inhibit its oligomerization into plasma membrane , suppressing pyroptosis in inflammatory models. MCC950, a potent NLRP3-specific , binds the NACHT domain's Walker B , blocking NLRP3's activity and preventing ASC oligomerization, which halts caspase-1-mediated GSDMD and pyroptotic . Post-translational modifications further fine-tune pyroptosis by stabilizing inactive protein conformations. O-GlcNAcylation of GSDMD at specific serine residues, such as Ser338, hinders its cleavage by inflammatory , reducing GSDMD-NT release and subsequent pore formation in endothelial cells during . Recent advances have identified palmitoylation of GSDMD-NT at Cys191 as a critical checkpoint for targeting; inhibitors like NU6300, which covalently react with this residue, stabilize inactive GSDMD forms, suppress pyroptosis in macrophages, and improve outcomes in septic models as demonstrated in 2024 studies. Negative feedback loops provide additional layers of control to limit pyroptosis and prevent excessive . IL-1 receptor (IL-1R) signaling, triggered by pyroptosis-derived IL-1β, induces expression of suppressors of signaling (SOCS) proteins, particularly SOCS1 and SOCS3, which inhibit further activation by promoting its ubiquitination and degradation, thereby downregulating the pyroptotic cascade. serves as a degradative that selectively targets components, such as and ASC specks, for lysosomal breakdown, thereby preventing caspase-1 activation and GSDMD-mediated pyroptosis in infected macrophages. Certain physiological triggers exhibit dual roles in pyroptosis regulation depending on concentration. Reactive oxygen species (ROS), while typically promoting NLRP3 activation at moderate levels, inhibit pyroptosis at high concentrations by oxidizing critical cysteines in caspase-1, such as Cys285, which impairs its autocatalytic activity and blocks GSDMD cleavage.

Physiological Roles

Role in Host Defense Against Pathogens

Pyroptosis serves as a critical component of innate immunity by enabling the lytic death of infected cells, which disrupts intracellular pathogen replication and amplifies inflammatory signals to orchestrate broader host defense. In bacterial infections, pyroptosis effectively restricts pathogens such as Salmonella typhimurium and Listeria monocytogenes through caspase-1-mediated cleavage of gasdermin D, leading to plasma membrane rupture that ejects bacteria into the extracellular space for subsequent elimination by neutrophils via reactive oxygen species. This process is particularly prominent in the spleen, where pyroptosis acts as the primary clearance mechanism against Gram-negative bacteria like Chromobacterium violaceum, reducing bacterial burden and preventing systemic dissemination. Concurrently, the release of interleukin-1β (IL-1β) and IL-18 from pyroptotic cells recruits and activates neutrophils and natural killer cells, enhancing local inflammation and pathogen containment without excessive tissue damage in neutrophils, which resist pyroptosis themselves. In viral infections, ZBP1 (Z-DNA binding protein 1) emerges as a key sensor that initiates pyroptosis to counter threats like (IAV) and (HSV-1). ZBP1 detects viral nucleic acids, such as IAV nucleoprotein and polymerase basic 1, or HSV-1 DNA, activating the inflammasome and caspase-1 to drive gasdermin D pore formation, thereby inducing inflammatory that limits viral propagation. This response promotes IL-1β and IL-18 secretion, bolstering antiviral immunity, though it represents a double-edged sword as viruses employ various evasion tactics to block ZBP1 signaling and prevent inflammatory . For fungal and parasitic pathogens, the AIM2 inflammasome detects cytosolic double-stranded DNA from microbes, including fungal elements and parasitic invaders, triggering caspase-1 activation that culminates in pyroptosis to curtail pathogen spread. This DNA-sensing mechanism restricts replication of DNA pathogens, such as certain fungal DNA mimics or parasites like , by eliminating infected host cells and releasing alarmins that amplify innate responses. The protective role of pyroptosis extends across evolutionary lineages, with gasdermin-mediated pore formation conserved from to mammals, underscoring its ancient origins in antimicrobial defense. In teleosts like , inflammasome components and gasdermins enable similar lytic responses to pathogens, highlighting functional preservation over 450 million years of divergence. Beyond immediate innate effects, pyroptosis enhances adaptive immunity by liberating pathogen-associated antigens from ruptured cells, which are captured by dendritic cells to prime T-cell responses and generate . Recent 2025 analyses further illuminate pyroptosis's context-dependent benefits in viral infections like , where early activation eliminates SARS-CoV-2-infected cells via NLRP3-gasdermin D pathways, releasing IL-1β and IL-18 to recruit antiviral effectors and control initial replication. As of 2025, studies highlight pyroptosis's role in defending against emerging zoonotic viruses via ZBP1-PANoptosis integration. However, unchecked pyroptosis can escalate to hyperinflammation, contributing to cytokine storms in severe cases, emphasizing the need for balanced regulation.

Involvement in Tissue Development and Homeostasis

Pyroptosis plays a pivotal role in embryonic by facilitating the clearance of damaged neural cells in the , thereby preventing the accumulation of damage and ensuring proper . During cortical , high levels of replicative stress induce damage in neural progenitors, triggering gasdermin D (GSDMD)-mediated pyroptosis via the AIM2 pathway. This process eliminates defective cells in the ventricular and subventricular zones, promoting balanced and of healthy progenitors. Disruption of this mechanism leads to excessive progenitor accumulation, impaired neuronal maturation, and long-term neurodevelopmental abnormalities, such as autism-like behaviors in mice. In tissue , pyroptosis contributes to maintaining epithelial barrier integrity in the gut by selectively removing stressed or senescent enterocytes without compromising overall tissue function. Controlled activation of pyroptosis in intestinal epithelial cells helps regulate microbial interactions and prevents barrier leakage, supporting mucosal through the timely clearance of damaged cells. This balanced inflammatory response is essential for tissue repair in various organs. Pyroptosis also regulates stem cell populations by pruning excess progenitors, particularly in hematopoiesis, where it maintains (HSC) balance under stress conditions. In HSCs, GSDME-mediated pyroptosis eliminates overproliferating or damaged cells in response to genotoxic agents like , preventing clonal expansion and preserving long-term repopulation capacity. Similar pruning mechanisms operate in skin stem cell niches, where pyroptosis of removes aberrant cells to sustain epidermal renewal and barrier function without disrupting tissue architecture. In non-inflammatory contexts, low-level GSDMD activation supports tissue remodeling by forming transient pores that enable release and cellular content extrusion without inducing full lytic or a . This sub-lytic pore formation facilitates coordinated cell extrusion in epithelial sheets, aiding in and developmental while preserving tissue integrity. Such regulated activity contrasts with robust pyroptosis, allowing pyroptosis to contribute to through subtle adjustments rather than overt . Animal models underscore these roles, with GSDMD knockout mice exhibiting developmental defects in the , including disrupted cortical layering and progenitor overaccumulation due to failed DNA damage clearance. In the gut, GSDMD deficiency impairs epithelial turnover, leading to barrier dysfunction and altered microbial , highlighting pyroptosis's necessity for normal tissue maturation. These phenotypes demonstrate that GSDMD-mediated pyroptosis is indispensable for preventing developmental anomalies and sustaining adult tissue equilibrium.

Pathological Implications

In Infectious Diseases

In bacterial infections, excessive activation of the leads to pyroptosis, contributing to severe pathology in conditions like . For instance, (LPS) from triggers NLRP3-dependent pyroptosis in endothelial cells, driving endotoxic shock through gasdermin D (GSDMD) pore formation and release of pro-inflammatory cytokines. This process amplifies , as miR-21 enhances NLRP3 inflammasome activity, promoting pyroptosis and via pathway dysregulation. In caused by , pyroptosis initially aids host defense by lysing infected macrophages and releasing IL-1β to recruit immune cells, but in chronic phases, excessive NLRP3 and AIM2 inflammasome activation causes tissue damage and facilitates bacterial dissemination. Viral infections also exploit or dysregulate pyroptosis for pathogenesis. In HIV-1 infection, pyroptosis of CD4+ T cells, mediated by caspase-1 cleavage of GSDMD, drives immune depletion and viral persistence by reducing antiviral responses, with HIV-1 proteins like Vpu contributing to through caspase-3/GSDME pathways in the brain. Similarly, induces ZBP1-dependent PANoptosis—a coordinated pyroptotic, apoptotic, and necroptotic —in lung epithelial cells, leading to with elevated IL-1β and TNF-α levels that exacerbate acute respiratory distress. In parasitic infections such as , AIM2 inflammasome activation by Plasmodium DNA triggers pyroptosis in infected cells, releasing IL-1β and IL-18 that intensify and exacerbate cerebral damage. Dual engagement of AIM2 and by hemozoin and parasite DNA further promotes , contributing to blood-brain barrier disruption and neuronal injury in cerebral malaria. As of 2025, pyroptosis exhibits a double-edged role in antibiotic-resistant infections, where it enhances clearance of resistant like species by restricting intracellular replication, but excessive activation worsens tissue damage in persistent infections. Plant-derived compounds, such as polyphenols and from and food sources, act as inducers of targeted pyroptosis in macrophages, boosting bacterial clearance while minimizing hyperinflammation in models of resistant infections. Dysregulated pyroptosis in these infections culminates in and IL-1β-mediated tissue damage, with elevated cytokines causing vascular leakage and organ failure. Animal models demonstrate that caspase-1 inhibitors, such as AC-YVAD-CMK, reduce mortality in LPS-induced by blocking GSDMD cleavage and IL-1β release, alleviating and injury.

In Neurovascular and Cardiovascular Diseases

Pyroptosis plays a in neurovascular diseases, particularly ischemic stroke, where the activates gasdermin D (GSDMD) to mediate inflammatory in neurons, , and endothelial cells. This process leads to blood-brain barrier () breakdown through caspase-1-dependent cleavage of GSDMD, forming membrane pores that release pro-inflammatory such as IL-1β and IL-18, thereby amplifying ischemic injury and promoting neuronal death. In mouse models of middle cerebral artery occlusion/reperfusion (MCAO/R), activation via ROS/TXNIP signaling exacerbates permeability and infarct expansion, while inhibition of this pathway with compounds like astragaloside IV reduces pyroptosis and decreases infarct volume, and improves neurological function. Similarly, GSDMD knockout in mice subjected to MCAO diminishes infiltration, lowers levels (e.g., IL-1β, IL-6, TNF-α), and reduces infarct size by approximately 30-50% at day 3 post-stroke, alongside enhanced sensory and motor recovery. In neurodegenerative conditions like (AD), amyloid-β (Aβ) peptides activate the AIM2 inflammasome in , triggering GSDMD-mediated pyroptosis that releases damage-associated molecular patterns (DAMPs) and cytokines, which in turn amplify tau hyperphosphorylation and aggregation. This creates a feed-forward loop of , where pyroptotic exacerbate plaque formation and neuronal loss, contributing to cognitive decline. Studies in APP/PS1 mouse models demonstrate that AIM2 or inhibition attenuates Aβ-induced microglial pyroptosis, reduces tau pathology, and lowers Aβ deposition, highlighting the pathway's role in disease progression. Emerging evidence also indicates sex differences, with female AD models showing elevated NLRP1 inflammasome signaling and greater pyroptotic susceptibility compared to males, potentially linked to hormonal influences on microglial activation. In cardiovascular diseases, GSDMD-driven pyroptosis contributes to by promoting death within plaques, leading to instability and rupture; -derived GSDMD perforates mitochondria, releasing mtDNA that activates the STING-IRF3/ axis and sustains inflammation. In ApoE^{-/-} mice on a high-fat , GSDMD reduces plaque area by over 50%, decreases content from 56% to 21%, and lowers IL-1β/IL-18 levels, confirming its role in plaque progression. For (MI), mitochondrial (mtROS) initiate NLRP3 assembly in cardiomyocytes, culminating in GSDMD pore formation and cell lysis during ischemia/reperfusion. Recent 2025 analyses underscore GSDMD's effector dominance in MI, where its conditional in cardiomyocytes shrinks infarct size and preserves cardiac function in murine models. Across these conditions, DAMPs such as and ATP, released from initial pyroptotic cells, bind pattern recognition receptors to propagate /AIM2 activation, establishing self-amplifying inflammatory cycles that worsen tissue damage. In both and MI animal models, GSDMD ablation interrupts this cycle, mitigating global inflammation and cell loss while improving outcomes like reduced cognitive impairment in AD or heart failure post-MI.

In Cancer

Pyroptosis exhibits a dual role in cancer, acting as both a tumor suppressor and promoter depending on the cellular context and triggering pathway. In its suppressive capacity, gasdermin E (GSDME)-mediated pyroptosis in chemotherapy-sensitive tumors releases damage-associated molecular patterns (DAMPs), such as and ATP, which activate dendritic cells and enhance + T-cell infiltration into the , thereby amplifying antitumor immune responses. This mechanism is particularly evident in tumors where chemotherapeutic agents like cleave GSDME via caspase-3, converting to immunogenic pyroptosis and improving therapeutic efficacy. Conversely, in promotive scenarios, inflammasome activation leading to pyroptosis in tumor-associated macrophages (TAMs) releases IL-1β and IL-18, which polarize additional macrophages toward an M2-like immunosuppressive phenotype, fostering tumor progression and immune evasion. High expression in TAMs correlates with poorer outcomes in cancers such as head and neck . Cell-type specificity further modulates pyroptosis's impact, with elevated gasdermin B (GSDMB) expression observed in epithelial cancers, including and , where it drives non-canonical pyroptosis via caspase-4/5 or granzyme A, potentially contributing to tumor but also that supports in certain contexts. Low GSDME expression, frequently downregulated in gastric, colorectal, and cancers through promoter hypermethylation, is associated with aggressive disease and poor prognosis, as it impairs pyroptotic responses to therapies and reduces immunogenic . In , for instance, diminished GSDME levels predict worse survival and radioresistance. Recent advances as of 2025 highlight plant-derived inducers, such as from sources like and , which activate or caspase-3/GSDME pathways to trigger pyroptosis in drug-resistant cancers, including multidrug-resistant ovarian and lung tumors, thereby restoring sensitivity and eliciting antitumor immunity with minimal toxicity. Targeting pyroptosis in contexts has shown promise in overcoming PD-1 resistance; for example, inducing GSDME-dependent pyroptosis enhances T-cell effector functions and reverses immunosuppressive barriers in models resistant to checkpoint blockade. Clinically, pyroptosis-related gene signatures, incorporating markers like GSDMD and , predict favorable responses to CAR-T in hematologic malignancies by indicating robust activity that boosts release and T-cell persistence. Mouse models further demonstrate that engineered pyroptosis induction, such as via -armed GSDME activation, significantly enhances anti-tumor immunity, leading to tumor regression in solid tumors like and promoting long-term T-cell memory.

In Metabolic and Autoimmune Disorders

Pyroptosis plays a significant role in metabolic disorders such as and , where activation of the in macrophages promotes and . In obese individuals, NLRP3-driven pyroptosis in visceral exacerbates the release of pro-inflammatory cytokines like IL-1β, contributing to systemic insulin desensitization and metabolic dysfunction. Recent studies have highlighted the IL-9–NLRP3 axis as a modulator in this process, with reduced IL-9 levels correlating with enhanced NLRP3 activity and worsened in patients. Similarly, in induces caspase-1 activation in pancreatic β-cells, triggering GSDMD-mediated pyroptosis and β-cell dysfunction, which impairs insulin secretion and accelerates disease progression. This mechanism is evidenced by high-glucose conditions promoting NLRP3/GSDMD signaling, leading to inflammatory cell death in β-cells. In , monosodium urate (MSU) crystals, formed from , directly induce pyroptosis in macrophages and neutrophils, perpetuating a cycle of and uric acid accumulation in joints. MSU crystals activate the , resulting in caspase-1 cleavage of GSDMD and subsequent pore formation, which amplifies IL-1β release and sustains acute flares. This pyroptotic response in synovial macrophages is critical for the pathological progression of , as inhibiting GSDMD reduces crystal-induced and . Autoimmune disorders, particularly cryopyrin-associated periodic syndromes (CAPS), arise from gain-of-function mutations in , leading to constitutive activation, excessive IL-1β production, and pyroptosis in affected tissues. These mutations cause uncontrolled caspase-1 activity, resulting in skin rashes, joint inflammation, and systemic autoinflammation characteristic of CAPS. Pathogenic NLRP3 variants enhance spontaneous assembly, driving pyroptotic and IL-1β-mediated flares without external triggers. Recent advancements as of 2025 underscore the involvement of gasdermin D (GSDMD) in non-alcoholic fatty liver disease (NAFLD) progression, where GSDMD-mediated pyroptosis in hepatocytes exacerbates and through NLRP3-caspase-1 signaling. Additionally, AIM2 activation has been linked to pyroptosis in models, such as experimental autoimmune encephalomyelitis (EAE), where AIM2 regulates microglial and neuronal pyroptosis, contributing to central nervous system . stresses, including mitochondrial dysfunction and endoplasmic reticulum stress, amplify pyroptosis in these metabolic contexts by potentiating activation and release, forming a vicious cycle of . Therapeutically, IL-1 blockers like , a recombinant IL-1 , effectively mitigate pyroptosis-driven flares in autoimmune conditions such as CAPS by neutralizing IL-1β and reducing -mediated . rapidly alleviates symptoms in NLRP3-related , highlighting its role in targeting downstream pyroptotic outcomes.

Therapeutic Targeting

Pharmacological Inhibitors

Pharmacological inhibitors of pyroptosis target key components of the pathway, including , gasdermin D (GSDMD), and , to mitigate excessive in various diseases. These agents primarily act by blocking assembly, GSDMD cleavage and pore formation, or activation, thereby preventing the release of pro-inflammatory cytokines like IL-1β and IL-18, as well as lytic . Small molecules and biologics in this class have shown promise in preclinical models, though clinical translation remains limited by specificity and delivery hurdles. NLRP3 inhibitors represent a major focus due to the central role of the in pyroptosis initiation. MCC950, a sulfonylurea-based , selectively inhibits by binding to its NACHT domain and blocking activity, thereby preventing ASC oligomerization and downstream caspase-1 activation without affecting other like AIM2 or NLRC4. In preclinical studies, MCC950 has demonstrated efficacy in models of cryopyrin-associated periodic syndromes (CAPS) by reducing IL-1β production and pyroptosis in patient-derived cells, and in models where it attenuates monosodium urate crystal-induced inflammation and death. Similarly, CY-09 inhibits activation by disrupting ASC oligomerization, showing neuroprotective effects in models of cerebral ischemia by limiting pyroptosis and cognitive deficits. Both compounds remain in preclinical stages for CAPS and , with ongoing efforts to optimize MCC950 analogs for better potency and reduced toxicity. As of 2025, inhibitors like dapansutrile have advanced to Phase III trials for acute , showing reduced inflammation without affecting . Direct targeting of GSDMD, the executor of pyroptosis, offers a downstream approach to block pore formation and cell lysis. Disulfiram, an FDA-approved inhibitor repurposed for pyroptosis, covalently modifies residues in the GSDMD N-terminal fragment (GSDMD-NT), preventing pore assembly on the plasma membrane and reducing IL-1β release in inflammatory models. Ac-FLTD-CMK, a peptide-based inhibitor derived from the GSDMD cleavage site, specifically blocks -1, -4, -5, and -11-mediated GSDMD processing (IC50 of 46.7 nM for -1), thereby inhibiting pyroptosis in macrophages and improving outcomes in and models. Recent advancements include 2025 developments in small-molecule GSDMD inhibitors, such as necrosulfonamide (NSA) derivatives and novel compounds like GI-Y1, which target GSDMD pores or cleavage to attenuate myocardial injury in , though these are still preclinical. Broad-spectrum caspase inhibitors, such as , indirectly suppress pyroptosis by inhibiting activity. VX-765, an oral of the selective VRT-043198, reduces IL-1β maturation, GSDMD , and pyroptotic in models of , , and , with demonstrated reductions in via PPARα upregulation. However, selectivity challenges arise, as VX-765 can partially inhibit and -7 at higher doses, potentially interfering with and complicating therapeutic use in tissues where both pathways coexist. Clinically, IL-1 pathway blockers like (IL-1 receptor antagonist) and (anti-IL-1β monoclonal antibody) indirectly dampen pyroptosis by neutralizing downstream cytokines, reducing inflammasome-driven inflammation without directly targeting upstream components. These biologics have been evaluated in Phase II trials for acute ischemic stroke as of 2025, showing safety, reduced , and potential in small randomized controlled trials, though larger efficacy studies are needed. Key challenges in developing pyroptosis inhibitors include off-target effects on , particularly with inhibitors that may promote unintended cell survival in tumors or chronic inflammation, and poor cytosolic delivery for intracellular targets like and GSDMD, limiting efficacy . Ongoing research emphasizes structure-based design to enhance specificity and formulations for targeted delivery.

Inducers and Immunotherapeutic Strategies

Pyroptosis can be induced through various endogenous and exogenous stimuli that activate inflammasomes or caspases, leading to the cleavage and oligomerization of gasdermin family proteins such as GSDMD and GSDME. Canonical pathways involve caspase-1 activation by NLRP3 or AIM2 inflammasomes in response to damage-associated molecular patterns (DAMPs) such as ATP or pathogen-associated molecular patterns (PAMPs) like double-stranded DNA (for AIM2), resulting in GSDMD pore formation and release of IL-1β and IL-18. Non-canonical induction occurs via caspase-4/5/11 sensing cytosolic lipopolysaccharide (LPS), while alternative routes include caspase-3-mediated GSDME cleavage triggered by apoptotic signals or granzyme B from cytotoxic lymphocytes. Pharmacological inducers encompass chemotherapeutic agents like cisplatin and paclitaxel, which activate caspase-3/GSDME in tumor cells, and natural compounds such as docosahexaenoic acid (DHA), which stimulates NLRP3 inflammasomes. Emerging nanomedicines, including gold nanoparticles delivering GSDMA3 N-terminal domains or mRNA-lipid nanoparticles encoding GSDMD N-terminal fragments, precisely target pyroptosis to enhance therapeutic efficacy. Reactive oxygen species (ROS) generators, such as photosensitizers (e.g., NIR-II AIEgens), and ion-overloading agents like Fe²⁺-loaded nanoparticles further promote gasdermin activation by disrupting cellular homeostasis. Immunotherapeutic strategies leverage pyroptosis induction to transform immunosuppressive tumor microenvironments into immunogenic ones by releasing tumor antigens, DAMPs (e.g., ), and that recruit and activate dendritic s, T cells, and natural killer cells. In cancer settings, combining pyroptosis inducers with inhibitors (ICIs) like anti-PD-1/ antibodies has shown synergistic effects; for instance, -induced GSDME pyroptosis in small-cell upregulates IL-12 signaling, enhancing ICI responsiveness and improving survival in preclinical models. CAR-T cell therapies exploit granzyme B-mediated GSDME cleavage to trigger pyroptosis in target tumors, amplifying release and T-cell infiltration while mitigating exhaustion, as demonstrated in models where GSDME overexpression boosted antitumor activity. Oncolytic viruses, such as parapoxvirus ovis, induce pyroptosis via /GSDMD activation, acting as in situ vaccines that prime adaptive immunity and synergize with ICIs in . Nanocarrier-based approaches, including scaffolds or ZIF-8 nanoparticles co-delivering GSDMD inducers and PD-1 blockers, have eradicated "cold" tumors in mouse models by sustaining pyroptotic signaling and preventing immune escape. Clinical trials, such as NCT03349710 evaluating nivolumab with in advanced cancers, underscore the translational potential of these strategies to overcome resistance to monotherapy.