v
Search
Advanced

Publications > Journals > Gene Expression> Article Full Text

  • OPEN ACCESS

Inflammasome Activation as a Key Driver of Acetaminophen-induced Hepatotoxicity: Mechanisms and Emerging Therapeutics

  • Narmadhaa Sivagurunathan  and
  • Latchoumycandane Calivarathan* 
 Author information 

Abstract

Acetaminophen (APAP) is one of the most commonly used analgesic and antipyretic medications and is generally considered safe at therapeutic doses. However, overdose remains a leading cause of acute liver failure, primarily characterized by centrilobular (zone 3) hepatic necrosis, oxidative stress, mitochondrial dysfunction, and sterile inflammation. The hepatotoxic effects of APAP are localized to the centrilobular region, where cytochrome P450 2E1 is highly expressed. Cytochrome P450 2E1 catalyzes the conversion of APAP to a toxic metabolite, N-acetyl-p-benzoquinone imine. During overdose, the liver’s detoxification capacity is overwhelmed and excess N-acetyl-p-benzoquinone imine binds to cellular proteins, initiating oxidative stress and mitochondrial injury that culminate in hepatocyte death. A central component of APAP-induced hepatotoxicity is the activation of innate immune responses, particularly via inflammasome pathways. Inflammasomes are cytosolic multiprotein complexes that detect cellular damage and trigger inflammation. Among these, the NOD-, LRR-, and pyrin domain-containing 3 (NLRP3) inflammasome plays a significant role in APAP-induced liver injury. Upon activation, the NLRP3 inflammasome promotes autocatalytic cleavage of procaspase-1 into its active form, caspase-1, which subsequently processes the pro-inflammatory cytokines pro-interleukin-1β and pro-interleukin-18 into their mature forms. These cytokines recruit additional immune cells and amplify liver inflammation, exacerbating tissue injury. Thus, the NLRP3 inflammasome serves as a key mechanistic link between the initial toxic insult and the ensuing inflammatory response in APAP hepatotoxicity. This review aimed to explore the molecular mechanisms underlying APAP-induced liver injury, particularly inflammasome activation, and evaluate the current and emerging therapeutic strategies.

Graphical Abstract

Keywords

Acetaminophen, Caspase-1, Drug-induced liver injury, Inflammasome, NOD-, LRR-, and pyrin domain-containing 3, NLRP3

Introduction

Acetaminophen (APAP), commonly known as paracetamol, is a widely used over-the-counter medication renowned for its analgesic and antipyretic properties. Despite its ubiquity and recognized safety when taken within recommended doses, APAP overdose remains a significant public health concern worldwide due to liver injury, posing a substantial burden on healthcare systems and contributing to a considerable number of hospitalizations and fatalities annually.1 APAP overdose frequently occurs due to various factors, including intentional or unintentional ingestion beyond recommended doses or accidental overdoses resulting from unawareness of the presence of APAP in various combination products. Furthermore, in individuals with a history of alcoholism, fasting, or liver disease, even therapeutic doses of APAP can cause liver injury.2 The accessibility and widespread availability of APAP contribute to its high prevalence as a cause of acute liver injury. APAP toxicity, whether resulting from a single overdose or repeated supratherapeutic ingestion, progresses through four phases: preclinical toxic effects (phase one), hepatic injury (phase two), hepatic failure (phase three), and recovery (phase four). Early intervention during the preclinical phase generally leads to full recovery with minimal liver damage. However, delayed treatment after hepatic injury presents a variable prognosis, and progression to hepatic failure carries a mortality rate of 20% to 40%.3

The hepatotoxic effects of APAP overdose are well documented and primarily arise from the metabolic conversion of APAP to reactive metabolites in the liver. Under normal conditions, APAP is metabolized via conjugation with sulfate and glucuronide, leading to its excretion in the urine. However, a small fraction of APAP undergoes oxidative metabolism by cytochrome P450 enzymes, predominantly cytochrome P450 2E1 (CYP2E1), to form N-acetyl-p-benzoquinone imine (NAPQI), a highly reactive and toxic intermediate. The subsequent detoxification of NAPQI occurs through its conjugation with glutathione (GSH), a crucial endogenous antioxidant in the liver. However, when APAP is ingested in excessive amounts, the capacity of GSH to neutralize NAPQI becomes overwhelmed, leading to depletion of hepatic GSH stores and accumulation of NAPQI, resulting in oxidative stress, mitochondrial dysfunction, and hepatocellular injury, ultimately culminating in acute liver injury and potential liver failure. The clinical manifestations of APAP overdose vary widely depending on the dose ingested, the time elapsed since ingestion, and individual patient factors.2

Early symptoms may include gastrointestinal disturbances such as nausea, vomiting, and abdominal pain, which may progress to more severe manifestations, including jaundice, hepatic encephalopathy, and coagulopathy in patients with advanced liver injury.4 While timely recognition and intervention are crucial for mitigating the adverse effects of APAP overdose, effective treatment options are available to manage hepatotoxicity and prevent progression to liver failure. Despite treatment, hepatotoxicity occurs in 12–13% of acute APAP overdoses, with 2–5% of cases progressing to liver failure and 0.2–0.5% resulting in fatalities.5,6 N-acetylcysteine (NAC), a precursor of GSH, serves as the cornerstone of therapy for APAP overdose by replenishing depleted GSH stores and enhancing the detoxification of NAPQI. The administration of NAC within the appropriate timeframe reduces the risk of hepatotoxicity and significantly improves patient outcomes.7 Despite advancements in understanding the pathophysiology of APAP overdose and the development of effective treatment strategies, a considerable proportion of cases still progress to hepatotoxicity, liver failure, and even mortality. This review is significant as it provides a comprehensive exploration of the role of inflammasomes in APAP-induced liver injury, a critical but often underemphasized aspect of the pathophysiology of this condition. While the hepatotoxic effects of APAP overdose are well documented, the intricate interplay between oxidative stress, cellular damage, and the inflammatory response requires further elucidation. The purpose of this review was to critically evaluate the molecular basis of APAP-induced liver injury, with a focus on inflammasome signaling and emerging therapeutic interventions. By incorporating recent advances, this work provides a comprehensive understanding of the role of cytosolic multiprotein complexes, particularly inflammasomes, in the molecular pathogenesis of liver injury. Furthermore, this review identifies potential therapeutic targets by elucidating the mechanisms of inflammasome activation and their downstream effects. Such information is crucial for developing novel treatments aimed at mitigating the inflammatory response and reducing liver damage in patients with APAP-induced liver injury. Additionally, this study underscores the need for clinical approaches that address both the toxicological aspects and modulation of the inflammatory response to improve patient outcomes.

Mechanistic pathways associated with acetaminophen-induced hepatotoxicity

APAP is rapidly absorbed and extensively metabolized upon entering the liver, with 85–90% conjugated with glucuronic acid or sulfate by the enzymes uridine diphosphate-glucuronosyltransferase and sulfotransferase and subsequently eliminated. This conjugation renders APAP metabolites water-soluble, aiding their excretion via urine or bile. By converting APAP into water-soluble forms, this phase II pathway helps detoxify and eliminate APAP from the body, reducing the risk of metabolite accumulation and potential harm.8 When APAP is administered at therapeutic doses under normal physiological conditions, the remaining 5–10% of the drug is metabolized in hepatocytes by cytochrome P450 enzymes such as CYP2E1 and CYP1A2 through phase I enzymatic reactions, resulting in the generation of the toxic reactive metabolite NAPQI (Fig. 1). Glutathione-S-transferase catalyzes the conjugation of NAPQI with GSH, forming APAP-GSH adducts,9,10 which are further metabolized into nontoxic thiolate and cysteine molecules, including adducts of cysteamide, glycine/cysteine, and N-acetyl-L-cysteine. Phase III involves the transport of these metabolites by transporters, after which they are eliminated through bile or urine, preventing potential adverse effects on the liver.

Acetaminophen-induced hepatotoxicity: Under normal physiological conditions, APAP undergoes detoxification in the liver through sulfation and glucuronidation.
Fig. 1  Acetaminophen-induced hepatotoxicity: Under normal physiological conditions, APAP undergoes detoxification in the liver through sulfation and glucuronidation.

A small fraction of APAP is converted by CYP2E1 into the highly reactive NAPQI, which is normally detoxified by GSH. However, under pathophysiological conditions, especially during APAP overdose, excessive NAPQI accumulates and binds to cellular proteins, also triggering oxidative stress, leading to hepatocellular damage and the release of damage-associated molecular patterns (DAMPs), contributing to further liver injury and inflammation. APAP, acetaminophen; CYP2E1, cytochrome P450 2E1; ER, endoplasmic reticulum; GSH, glutathione; NAPQI, N-acetyl-p-benzoquinone imine; ROS, reactive oxygen species.

However, following APAP toxicity, endogenous glucuronide and sulfate cofactors become depleted, leading to increased concentrations of NAPQI and rapid depletion of cellular GSH. Consequently, the accumulated NAPQI binds covalently to the cysteine, methionine, tyrosine, and tryptophan residues of various proteins, forming NAPQI-protein adducts within the cell that disrupt normal physiological protein functions.11,12 NAPQI binds to housekeeping proteins, glutathione peroxidase (GPx),13 and adenosine triphosphate (ATP) synthase,14,15 among other proteins often abundant in mitochondria.12 Free radicals are generated when NAPQI oxidizes thiol groups in proteins.16 Furthermore, NAPQI binds to complexes I and II, disrupting the electron transport chain and allowing electrons to escape and produce superoxide radicals.17 Complex II is more susceptible to NAPQI inhibition than complex I.17 The formation of peroxynitrite occurs when superoxide radicals interact with nitric oxide, resulting in nitrotyrosine adducts within mitochondria. NAPQI binding modifies the α-subunit of mitochondrial ATP synthase, reducing ATP production.14 Therefore, peroxynitrite and reactive oxygen species (ROS) damage mitochondrial DNA, impair cellular respiration and ATP synthesis, and ultimately cause mitochondrial dysfunction (Fig. 2). Mitochondrial enzymes, such as GPx, catalase, and manganese superoxide dismutase 2, frequently neutralize free radicals, including superoxide anions and hydrogen peroxide. Thioredoxin (Trx), present in the cytosol as Trx1 and in mitochondria as Trx2, is oxidized and upregulated in response to oxidative stress and elevated ROS levels.18 The phosphorylation of apoptosis signal-regulating kinase 1 (ASK1) is triggered by the dissociation of ASK1 from Trx, resulting in ASK1 activation (Fig. 2). Activated ASK1 phosphorylates c-Jun N-terminal kinase (JNK) via cytosolic mitogen-activated protein kinase 4/7.19 Following further translocation to the mitochondria, activated phosphorylated JNK initiates a cascade in which JNK binds to the outer mitochondrial membrane scaffold protein Sab, inhibiting electron transport and thereby increasing the generation of ROS and peroxynitrite.20

Mitochondrial dysfunction: NAPQI binds to mitochondrial proteins, disrupting ATP synthesis and increasing ROS production.
Fig. 2  Mitochondrial dysfunction: NAPQI binds to mitochondrial proteins, disrupting ATP synthesis and increasing ROS production.

Oxidative stress activates the ASK1-JNK pathway, leading to JNK translocation to mitochondria, further exacerbating mitochondrial damage. This results in membrane instability and the release of DAMPs, triggering inflammation and hepatocyte injury. ASK1, apoptosis signal-regulating kinase 1; ATP, adenosine triphosphate; DAMPs, damage-associated molecular patterns; GSH, glutathione; JNK, c-Jun N-terminal kinase; NAPQI, N-acetyl-p-benzoquinone imine; ROS, reactive oxygen species.

Furthermore, phosphorylated JNK induces oxidative stress, mitochondrial membrane permeability, and dysfunction through the activation of Bax, the translocation of Bax to the mitochondria, the opening of the mitochondrial permeability transition pore, and Src-dependent inhibition of the electron transport complex.12,21,22 The opening of the mitochondrial permeability transition pore under pathological conditions, such as oxidative stress, calcium overload, or energy depletion, leads to disruption of the mitochondrial membrane potential and the release of proapoptotic factors, ultimately culminating in cell death.23 Some of the proteins released from mitochondria into the cytoplasm include apoptosis-inducing factor and endonuclease G, both of which have nuclear localization signals and thus translocate to the nucleus, where the endonuclease cleaves DNA, resulting in DNA fragmentation.24,25 Similarly, apoptosis-inducing factor in the nucleus causes chromatin condensation and DNA fragmentation.12,26 Furthermore, the formation of NAPQI adducts contributes to oxidative stress, mitochondrial dysfunction, disrupted Ca2+ homeostasis, necrotic cell death, centrilobular necrosis, and eventual liver failure (Figs. 3 and 4). Cellular swelling, karyolysis, karyorrhexis, vacuolization, release of cellular contents, endoplasmic reticulum stress, sterile inflammation, and autophagy are other mechanisms associated with APAP hepatotoxicity.27

Hepatocyte necrosis: apoptosis-inducing factor (AIF) and endonuclease G, along with other mitochondrial components, are released into the cytoplasm, causing chromatin condensation and DNA fragmentation.
Fig. 3  Hepatocyte necrosis: apoptosis-inducing factor (AIF) and endonuclease G, along with other mitochondrial components, are released into the cytoplasm, causing chromatin condensation and DNA fragmentation.

This ultimately leads to hepatocyte necrosis, triggering the release of DAMPs. DAMPs, damage-associated molecular patterns.

Inflammasome-mediated liver injury: DAMPs released from stressed hepatocytes, damaged mitochondria, and necrotic liver cells activate inflammasome sensors, triggering the oligomerization and activation of inflammasome components.
Fig. 4  Inflammasome-mediated liver injury: DAMPs released from stressed hepatocytes, damaged mitochondria, and necrotic liver cells activate inflammasome sensors, triggering the oligomerization and activation of inflammasome components.

This process leads to caspase-1 activation, which facilitates the maturation and release of IL-18 and IL-1β. These proinflammatory cytokines recruit macrophages and neutrophils, intensifying inflammation and exacerbating liver injury, ultimately progressing to acute liver failure. IL, interleukin; DAMPs, damage-associated molecular patterns.

Studies have identified p53, a tumor suppressor protein, as a protective factor against APAP-induced liver toxicity. In animal models of APAP overdose, inhibition of p53 has been shown to worsen liver injury, indicating that p53 plays a crucial role in mitigating the damage caused by APAP.28 By activating p53, the liver initiates processes that promote cell cycle arrest, DNA repair, and apoptosis in damaged cells,29 helping to limit the extent of liver injury and facilitating the removal of cells that have sustained significant damage. Inhibition of p53 disrupts these protective cellular mechanisms, allowing APAP-induced damage to progress unchecked, resulting in severe liver injury.28 NAPQI has been shown to modify cysteine residues of the Kelch-like ECH-associated protein 1 (Keap1) protein, activating the Keap1-nuclear factor erythroid 2-related factor 2 (Nrf2) pathway.30 Keap1 regulates the transcription factor Nrf2 by promoting its ubiquitination and degradation under normal conditions. During oxidative stress, Keap1 undergoes modifications that inhibit its interaction with Nrf2, allowing Nrf2 to accumulate and activate the expression of antioxidant and cytoprotective genes. This enhances cellular defense against oxidative damage and inflammation,31 playing a crucial role in APAP-induced liver injury. When modified by NAPQI, Keap1 releases Nrf2, enabling its translocation into the nucleus, where it binds to antioxidant response elements in DNA and promotes transcription of genes encoding various antioxidant enzymes. These enzymes play a crucial role in combating NAPQI-induced oxidative stress by scavenging ROS and restoring cellular redox balance.30,32 Consequently, activation of the Keap1-Nrf2 pathway serves as a defense mechanism to strengthen the antioxidant system and minimize the detrimental effects of oxidative stress in the liver caused by NAPQI.30

Acetaminophen-induced necroptosis and inflammation

The mechanism of cell death following APAP overdose has been the subject of extensive investigation for several decades. Early studies proposed necrosis as the predominant mode of cell death, based on hallmark features including karyorrhexis, karyolysis, plasma membrane rupture, and a robust inflammatory response.33,34 However, evidence for apoptosis has only been partially validated, as it lacks both caspase activation and the formation of apoptotic bodies.27,35–37 Moreover, despite the efficacy of potent caspase inhibitors, they failed to prevent APAP-induced cell death.37

Necroptosis is a regulated form of necrotic cell death initiated by the binding of tumor necrosis factor (TNF)-α to its receptor TNFR1, forming a multiprotein signaling complex.38 Under conditions where caspase-8 activity is inhibited or absent, receptor-interacting protein kinase 1 (RIPK1) interacts with RIPK3 to form the necrosome complex. Within this complex, RIPK3 is phosphorylated, which subsequently activates mixed lineage kinase domain-like protein (MLKL). Phosphorylated MLKL oligomerizes and translocates to the plasma membrane, disrupting membrane integrity and ultimately resulting in cell lysis and inflammation. RIPK3 levels have been shown to be elevated in APAP-overdosed mice39; however, MLKL knockout mice remained susceptible to APAP-induced liver injury, indicating that necroptosis is not the primary mode of cell death in APAP-induced toxicity.34,40,41

Ferroptosis, another form of regulated cell death characterized by iron-dependent accumulation of ROS, lipid peroxidation, and oxidative membrane damage, is increasingly recognized as a key contributor to APAP-induced liver injury.42 Ferroptosis is initiated by the depletion of GSH, leading to the inactivation of GPx4, a key enzyme that protects against lipid peroxidation. In the absence of GPx4 activity, lipid peroxides accumulate within cellular membranes. Simultaneously, ferrous iron (Fe2+) reacts with hydrogen peroxide (H2O2) via the Fenton reaction to generate highly reactive hydroxyl radicals (OH), which further propagate lipid peroxidation. This oxidative damage to membrane lipids compromises membrane integrity and ultimately results in cell death.40 This mechanistic understanding highlights potential therapeutic avenues targeting ferroptosis for mitigating APAP-induced liver injury.43 Ferroptosis inhibitors, such as ferrostatin-1 and UAMC-3203, were assessed for their ability to attenuate APAP hepatotoxicity. UAMC-3203 only partially attenuated liver injury by inhibiting the translocation of JNK protein and subsequently reducing mitochondrial damage and liver injury, while ferrostatin-1 demonstrated no protective effect. Hence, ferroptosis is likely not a major contributing factor in APAP-induced hepatotoxicity under physiological conditions.44

Pyroptosis has been reported as a significant mechanism of cell death in APAP-induced liver injury, involving activation of the NLRP3 inflammasome and subsequent caspase-1 activation. This cascade results in the cleavage of Gasdermin D, leading to the formation of membrane pores that may contribute to cell lysis and the release of the proinflammatory cytokines interleukin (IL)-1β and IL-18.45–47 However, recent investigations have raised questions about the exclusive role of pyroptosis in APAP-induced hepatocyte loss.48 Studies using Gasdermin D knockout models have demonstrated partial protection against hepatocyte loss, while pancaspase inhibitors failed to prevent cell death, although they reduced IL-1β production.34,49,50 These findings suggest that APAP-induced cell death may not strictly conform to conventional definitions of pyroptosis or other established modes of cell death, highlighting the complexity and distinct characteristics of liver injury induced by APAP.

During APAP overdose, damaged hepatocytes release damage-associated molecular patterns (DAMPs) such as high mobility group box-1 (HMGB1), ATP, and DNA fragments. These DAMPs activate the innate immune system, particularly Kupffer cells (liver-resident macrophages) and recruited monocytes, which release proinflammatory cytokines like TNF-α, IL-1β, and IL-6.51 These cytokines amplify the inflammatory response. While the adaptive immune response is less immediate, there is evidence that it can be activated in the later stages of APAP-induced liver injury, contributing to sustained inflammation and liver damage.52

DAMPs, which include nuclear DNA fragments, ATP, HMGB1 protein, and mitochondrial DNA, are released into the bloodstream and recruit inflammatory cells to the site of liver injury. These include macrophages, neutrophils, natural killer cells, and natural killer T cells, which help clear necrotic cell debris and aid in liver cell recovery.53,54 However, it has also been suggested that the enhanced release of proinflammatory cytokines (TNF-α, IL-1β, IL-6, and IL-10) and chemokines (monocyte chemoattractant protein-1, macrophage inflammatory protein-2, and IL-8) further promotes hepatocyte damage, thereby aggravating necrosis.55 APAP-induced hepatocyte death triggers a neutrophilic inflammatory response, contributing to the worsening of existing injury.56 Studies have also validated the role of neutrophils in the pathophysiology of APAP hepatotoxicity and the release of IL-1β, which is produced when caspase-1 cleaves pro-IL-1β.56,57 The pathophysiological significance of this reaction to APAP is still controversial; whether it provides cellular protection against toxicity or worsens liver damage remains unclear. APAP-induced hepatotoxicity occurs in distinct phases, with inflammation acting as a promoter during the early stages. In later phases, inflammation facilitates the clearance of dead cells and cellular debris, thereby contributing to the stimulation of liver regeneration and repair.58

Inflammasome activation and its role in inflammatory responses

The inflammasome, a multiprotein complex, plays a central role in innate immunity and inflammatory responses by detecting cellular stress and danger signals, thereby coordinating the initiation of inflammatory pathways in response to diverse stimuli. The inflammasome complex comprises several key components, including a sensor, an adaptor, and an effector protein.59 Inflammasomes are classified based on the sensor proteins that initiate their oligomerization. Some of the major types of inflammasomes characterized to date include NOD-, LRR-, and PYD domain-containing protein 3 (NLRP3), NOD-like receptor (NLR) family caspase activation and recruitment domain (CARD) domain-containing protein 4, absent in melanoma 2 (AIM2), NLRP1, NLRP6, and pyrin inflammasomes.60 Each inflammasome type has a specific sensor protein and can respond to different stimuli, contributing to the regulation of inflammatory responses and immune homeostasis under various physiological and pathological conditions.61

The sensor protein recognizes specific danger signals or pathogen-associated molecular patterns and undergoes conformational changes upon activation.60 The activated sensor protein recruits the adaptor protein, which acts as a scaffold for assembling the inflammasome complex. Several families of sensor proteins, including NLRs, AIM2-like receptors, and pyrin, are involved in forming inflammasome complexes. Each sensor protein recognizes specific ligands and undergoes conformational changes upon ligand binding to initiate inflammasome assembly.61 The most commonly studied adaptor protein is apoptosis-associated speck-like protein containing a CARD (ASC), which contains a CARD that interacts with the CARD domain of pro-caspase-1.62 The effector protein is typically pro-caspase-1, an inactive precursor of caspase-1. Pro-caspase-1 is recruited to the complex upon inflammasome assembly and undergoes autoproteolytic cleavage to generate active caspase-1.60 Their primary function is to detect molecules known as DAMPs and pathogen-associated molecular patterns.63 By recognizing these signals, inflammasomes play a crucial role in initiating and regulating the inflammatory response in the body, particularly in response to cellular damage or microbial threats.61 Finally, the effector protein, typically a caspase enzyme, is recruited to the complex and activated, initiating downstream signaling events that lead to inflammation and cell death. One well-characterized inflammasome complex is the NLR family, which includes the prototypical NLRP3 inflammasome. NLRP3 senses a wide array of endogenous and exogenous danger signals, including microbial products, environmental toxins, and host-derived DAMPs, making it a central player in inflammatory responses.64 Inflammasome complex activation is a tightly regulated process involving multiple steps. First, the sensor protein within the inflammasome complex recognizes specific danger signals or stressors, triggering its activation. This induces a conformational change in the sensor protein, allowing it to recruit the adaptor protein and initiate the assembly of the inflammasome complex. Once assembled, the inflammasome complex serves as a platform for activating caspase enzymes, particularly caspase-1.65 Activated caspase-1 cleaves the proinflammatory cytokines pro-IL-1β and pro-IL-18 into their active forms, promoting inflammation and immune responses.65,66 Additionally, caspase-1 activation leads to programmed cell death known as pyroptosis, characterized by cell swelling, membrane rupture, and the release of proinflammatory intracellular contents.63

Role of inflammasome activation in APAP-induced liver injury

Inflammasome activation has been implicated in the pathogenesis of various hepatic and systemic inflammatory diseases, including nonalcoholic fatty liver disease,67–69 alcoholic liver disease,70,71 viral hepatitis,72 and sepsis-induced liver injury.73 During pathogenesis, inflammasomes are activated in response to damage-associated molecular patterns and pathogen-associated molecular patterns, releasing proinflammatory cytokines such as IL-1β and IL-18.59 Furthermore, chronic inflammasome activation has been linked to metabolic disorders such as obesity and type 2 diabetes, contributing to systemic inflammation and hepatic dysfunction.74 The pathophysiology of APAP-induced liver injury involves a complex interplay of cellular events, with inflammasome activation emerging as a critical component of the inflammatory response.8 APAP overdose initiates a cascade of events within the liver. Under normal conditions, NAPQI is detoxified by conjugation with GSH. However, during APAP overdose, the excessive production of NAPQI overwhelms the cellular antioxidant capacity, leading to the accumulation of NAPQI-protein adducts.5 This process induces oxidative stress and mitochondrial dysfunction, resulting in hepatocyte necrosis and the release of cellular contents, including DAMPs. During the early stages of APAP-induced liver injury, stressed and damaged hepatocytes release various endogenous DAMPs, such as HMGB1,75 nuclear DNA fragments,76 uric acid,77 mitochondrial DNA,76 ATP, and heat shock protein-70, an inducible stress response protein that is translocated to the cell surface and released during cellular stress or necrotic death.78 These DAMPs play a major role in promoting neutrophil infiltration by activating antigen-presenting cells, including Kupffer cells and dendritic cells, which amplifies the immune response and the progression of liver injury.78 These DAMPs act as danger signals that activate innate immune receptors, including pattern recognition receptors. Among these receptors, NLRP3 plays a central role in sensing cellular stress and danger signals.79 NLRP3 activation requires two distinct signals: priming and activation. The priming signal involves the upregulation of NLRP3 expression and the synthesis of proinflammatory cytokines such as TNF-α and IL-1β, typically mediated by nuclear factor kappa B activation in response to inflammatory stimuli.80 Several DAMPs, including ATP, mitochondrial DNA, and ROS, serve as activation signals (Fig. 3). The NLRP3 inflammasome is a critical component of the innate immune system responsible for detecting cellular stress and damage signals. The upstream and downstream regulators of NLRP3 in APAP-induced liver injury are summarized in Tables 1 and 2.48,81–124

Table 1

Overview of the upstream regulators of NLRP3 in APAP-induced liver injury

Upstream regulatorsMechanism of action
Reactive oxygen species (ROS)
Mitochondrial ROSAPAP metabolism generates ROS from mitochondria, directly activating NLRP382,83
CYP2E1 activityCytochrome P450 2E1 metabolizes APAP into reactive intermediates, increasing oxidative stress and ROS84,85
Mitochondrial dysfunction
Mitochondrial damageAPAP overdose induces mitochondrial damage and mtDNA release, acting as DAMPs to activate NLRP38688
mPTP openingMitochondrial permeability transition pore opening releases proapoptotic factors, promoting dysfunction and NLRP3 activation8991
Ion flux
Potassium effluxAPAP-induced cellular damage causes potassium efflux, triggering NLRP3 activation81,91
Calcium signalingDisruptions in calcium homeostasis further promote NLRP3 activation92,93
Lysosomal destabilization
Lysosomal damageOxidative stress from APAP causes lysosomal membrane permeabilization, releasing cathepsins to activate NLRP394,95
High-mobility group box 1 (HMGB1)
HMGB1 releaseNecrotic hepatocytes release HMGB1, a DAMP that activates TLRs and subsequently NLRP39698
ATP release
Extracellular ATPDamaged hepatocytes release ATP, activating P2X7 receptor, leading to potassium efflux and NLRP3 activation99,100
Cytokines and chemokines
IL-1β and TNF-αIL-1β and TNF-α amplify the inflammatory response and upregulate NLRP3 via NF-κB signaling101
NADPH oxidase
NADPH oxidase activityContributes to ROS production, promoting NLRP3 activation102,103
MicroRNAs
miR-223Downregulation of miR-223, which suppresses typically NLRP3, enhances inflammasome activation104,105
Transcription factors
NF-κBUpregulates NLRP3 and pro-IL-1β expression in response to inflammatory stimuli101,106,107
STAT3Modulates the expression of inflammasome components, influencing NLRP3 activity108
Table 2

Overview of the downstream regulators of NLRP3 in APAP-induced liver injury

Downstream regulatorsMechanism of action
Caspase-1 activation
Caspase-1Activated by NLRP3 inflammasome, leading to the cleavage and activation of proinflammatory cytokines IL-1β and IL-18109,110
Proinflammatory cytokines
IL-1βCleaved and activated by caspase-1, leading to the recruitment of inflammatory cells and amplification of the inflammatory response111,112
IL-18Also cleaved and activated by caspase-1, contributing to the inflammatory response and immune cell recruitment112
Pyroptosis
Gasdermin DCleaved by activated caspase-1, forming pores in the cell membrane that lead to pyroptosis, a form of inflammatory cell death113115
Recruitment of immune cells
Neutrophils and macrophagesIL-1β and IL-18 promote the recruitment and activation of neutrophils and macrophages, exacerbating liver inflammation and damage116
Inflammatory mediators
HMGB1 releaseFurther amplifies inflammation by acting as a danger-associated molecular pattern117,118
Tissue damage and fibrosis
Hepatocyte deathPyroptosis and inflammatory cell infiltration lead to hepatocyte death and liver tissue damage48,119
FibrogenesisChronic inflammation can stimulate hepatic stellate cells, leading to fibrosis120,121
Systemic inflammatory response
Acute phase responseElevated levels of proinflammatory cytokines can trigger a systemic inflammatory response, affecting multiple organs122
Regulation of adaptive immunity
Dendritic cell activationIL-1β and IL-18 can enhance dendritic cell maturation and antigen presentation, linking innate and adaptive immunity123,124

Upon sensing the activation signal, NLRP3 recruits the adaptor protein ASC and pro-caspase-1, leading to inflammasome complex assembly. This multimeric complex facilitates the autocatalytic activation of caspase-1, an inflammatory caspase, which cleaves pro-IL-1β and pro-IL-18 into their mature, biologically active forms, IL-1β and IL-18. These cytokines are potent mediators of inflammation and play crucial roles in orchestrating innate and adaptive immune responses.79 The release of mature IL-1β and IL-18 amplifies the inflammatory response in the liver, leading to the recruitment and activation of immune cells, including neutrophils, monocytes, and macrophages. These cells produce additional proinflammatory mediators, such as TNF-α, IL-6, and ROS, further exacerbating liver inflammation and hepatocyte injury. The sustained inflammatory cascade ultimately contributes to hepatocyte death, liver dysfunction, and progression to acute liver failure in severe cases of APAP overdose (Fig. 4).

Inflammasomes are reported to be involved primarily in sterile inflammation, promoting the removal of necrotic cellular debris, hepatocyte proliferation, and liver tissue regeneration.54 A recent study demonstrated that cross-talk between necroptosis and the NLRP3 inflammasome promotes APAP-induced liver injury.125 Targeting inflammasomes in APAP-induced liver injury has emerged as a potential therapeutic strategy to mitigate inflammation, tissue damage, and liver injury. NLRP3 activation in the liver results in shortened survival, hepatocyte pyroptosis, severe liver inflammation, and neutrophil infiltration.46,125,126 NLRP3−/−, Casp1−/−, and ASC−/− mice are less susceptible to APAP-induced liver injury and show reduced mortality.57 Genetic ablation of NLRP3 significantly reduces the release of cleaved IL-1β and IL-18, thereby alleviating APAP-induced liver injury.57 Similarly, inhibiting NLRP3 by suppressing Toll-like receptors (TLRs) 4 and 9 signaling pathways also results in hepatoprotection against APAP-induced liver injury.57,127,128 Suppressing NLRP3 inflammasome activation, ASC speck formation, caspase-1 activation, and IL-1β maturation alleviates acute liver injury caused by APAP poisoning.129–131 The administration of pharmacological inhibitors targeting inflammasome components, such as NLRP3 or caspase-1 inhibitors, mitigates APAP-induced liver injury in preclinical models, highlighting the pivotal role of inflammasomes in modulating hepatic inflammation and damage.132

Several phytocompounds, such as Sapidolide A,47 allicin,133 and berberine,134 have been shown to inhibit inflammasome activation and alleviate APAP-induced hepatotoxicity. Additionally, oridonin, identified as a covalent inhibitor of inflammasomes, has demonstrated significant efficacy in mitigating damage resulting from APAP overdose.135 AIM2 deficiency, another sensor involved in inflammasome activation, enhances APAP-induced inflammation and acute liver injury in aged mice by promoting oxidative stress and phosphorylation of JNK.136 Experimental models provide compelling evidence connecting inflammasome activation to APAP-induced liver injury. However, the role of inflammasomes in this injury remains unclear, as conflicting findings suggest inflammasomes may exacerbate liver injury. Endogenous IL-1β has been reported to be insufficient for activating and recruiting neutrophils, and even high pharmacological doses of IL-1β fail to exacerbate APAP-induced liver injury, indicating that IL-1 signaling is not a critical contributor to APAP-induced hepatotoxicity.49 Moreover, administration of high doses of IL-1β fails to exacerbate APAP-induced liver injury, further suggesting a limited role of IL-1 signaling in the pathogenesis of APAP hepatotoxicity.49,137 Consistently, IL-1 receptor-deficient (IL-1R−/−) mice show no significant protection following APAP overdose.49 Similarly, genetic ablation of NLRP3 provides no evidence of hepatoprotection.138 Inhibition of IL-1β maturation using pan-caspase inhibitors reduces circulating IL-1β levels without attenuating neutrophil infiltration, inflammation, or liver injury.48

Mice deficient in ASC, NLRP3, or caspase-1 fail to demonstrate significant protection against APAP-induced liver injury. The extent of hepatic damage, along with levels of proinflammatory cytokines, chemokines, and neutrophil infiltration, remains comparable to that observed in wild-type controls.137 Although recent studies support a role for NLRP3 inflammasomes in APAP-induced liver injury, the discrepancies and complexity of available data underscore the need for further research. Future investigations should focus on understanding the role of NLRP3 at different phases of APAP-induced liver injury, its activation in various cell types, including hepatocytes and Kupffer cells, and validating its involvement in liver damage in multiple animal models through comparative analyses. Further research is necessary to assess the therapeutic potential of NLRP3 inhibitors, particularly when combined with other medications.

Therapeutic implications

Therapeutic strategies for APAP-induced liver injury involve mitigating inflammation, preventing hepatocyte death, and promoting liver regeneration. Several approaches have been explored, including targeting inflammasome activation, inhibiting caspase-1, employing anti-inflammatory agents, and modulating the gut microbiota and bile acid metabolism. Inhibition of the NLRP3 inflammasome represents a promising therapeutic strategy for attenuating inflammation and reducing tissue damage in APAP-induced liver injury. Various small-molecule inhibitors targeting different stages of NLRP3 activation have been developed and investigated preclinically; these are listed in Table 3.47,131,133,139–149 These inhibitors may target upstream signaling events, such as potassium efflux,81 mitochondrial dysfunction,150 or lysosomal destabilization,151,152 all critical for NLRP3 activation. Small-molecule inhibitors such as MCC950 have shown efficacy in inhibiting NLRP3 activation and ameliorating liver injury in experimental models of APAP overdose.140 Scopoleitin mitigates APAP-induced hepatotoxicity through suppression of NLRP3 gene expression and concurrent inhibition of the Nrf2/HMGB1/TLR4/NF-κB signaling pathway, resulting in reduced levels of proinflammatory factors.141 Piperine exhibits a prophylactic effect against APAP-induced liver injury by modulating inflammasome activation, reducing oxidative stress and cellular necrosis, and ultimately limiting activation of pro-IL-1β.142 Aloperine inhibits HMGB1/TLR4/NF-κB and NLRP3 inflammasome activation, ameliorating oxidative stress and inflammation, thereby mitigating APAP-induced liver injury.143 Salidroside downregulates the levels of NLRP3, ASC, caspase-1, and IL-1β and significantly protects the liver from APAP overdose in C57BL/6 mice.144 Similarly, Pien Tze Huang, a traditional Chinese medicine, also exhibits protective effects against APAP-induced liver injury by significantly reducing levels of NLRP3 inflammasomes and proinflammatory cytokines.145 Rosmarinic acid demonstrates a hepatoprotective role through inhibition of Nrf2-mediated downregulation of the NIMA (never in mitosis gene a)-related kinase 7-NLRP3 pathway.146 Sapidolide A, extracted from a folk medicine used in China to treat inflammatory diseases, inhibits expression and activation of the NLRP3 inflammasome when used as a pretreatment in a mouse model of APAP-induced liver injury.47 Emodin exerts anti-inflammatory effects by inhibiting the NLRP3 pathway, thereby preventing hepatotoxicity induced by APAP.147 Peroxiredoxin 3 modulates NLRP3-induced pyroptosis by regulating mitochondrial oxidative stress and inflammation, mitigating APAP-induced liver injury.148 Kaempferol inhibits HMGB1 and TLR4 and downregulates NLRP3 expression, reducing proinflammatory cytokine levels and preventing APAP hepatotoxicity.131 Similarly, sinomenine exhibits hepatoprotective effects through suppression of NLRP3 inflammasome activation, reduction of oxidative stress, and inhibition of the HMGB1/TLR4/NF-κB pathway.153 Allicin also demonstrates hepatoprotective effects against APAP-induced liver injury by attenuating oxidative stress and inhibiting the NLRP3 inflammasome pathway, effectively preventing the release of proinflammatory cytokines.133 Caspase-1 is a key effector molecule downstream of inflammasome activation and plays a central role in processing and releasing proinflammatory cytokines, such as IL-1β and IL-18. Pharmacological inhibitors targeting caspase-1 activity have been investigated for their potential to dampen the inflammatory response and reduce tissue damage in APAP-induced liver injury. In preclinical studies, compounds such as VX-765 and pralnacasan have demonstrated efficacy in inhibiting caspase-1 activation and protecting against inflammatory diseases.154 However, further research is needed to evaluate their safety and efficacy in clinical settings. Anti-inflammatory agents, such as corticosteroids and nonsteroidal anti-inflammatory drugs (NSAIDs), have been explored for their potential to attenuate inflammation and mitigate liver injury in APAP overdose.155,156 While corticosteroids have shown limited efficacy and potential adverse effects in clinical trials, NSAIDs, particularly those with selective cyclooxygenase-2 inhibition, have demonstrated some protective effects in experimental models. However, the use of NSAIDs in APAP overdose remains controversial due to concerns about exacerbating hepatotoxicity and gastrointestinal bleeding. Similarly, cyclosporin A, a well-characterized immunosuppressive agent, has demonstrated protective effects against APAP-induced hepatotoxicity when administered in combination with NAC in murine models.157 While corticosteroids and other immunosuppressive agents exert broad anti-inflammatory effects, direct inhibition of NLRP3 inflammasome activation represents a more specific therapeutic strategy, potentially allowing for more effective suppression of inflammation with reduced off-target effects.

Table 3

NLRP3 inhibitors and their mechanism of action against acetaminophen-induced liver injury

CompoundsMechanism of action
ScopoleitinAmeliorates the AILI through HMGB1/TLR4/NLRP3 inflammasome pathway141
PiperineProtects the liver from AILI by modulating the inflammasome pathway142
AloperineReduced oxidative stress and inflammation through the inhibition of HMGB1/TLR4/NF-κB and NLRp3 inflammasome pathway143
SalidrosideDownregulates the NF-κB/ NLRp3 inflammasome axis, thereby protecting the liver from acetaminophen-induced hepatotoxicity144
Pien Tze HuangInhibits NLRP3 inflammasome and protects the liver from damage145
Rosmarinic acidSuppresses the APAP-induced NEK7-NLRP3 activation and regulates the Nrf2-mediated inhibition of ROS production146
Sapidolide ARegulates the NLRP3 activation in macrophages and exerts anti-inflammatory effects47
EmodinUpregulates Nrf2-mediated antioxidative pathway and inhibits NLRP3 activation147
Peroxiredoxin 3Targets mitochondrial oxidative stress and inhibits NLRP3 activation148
KaempferolInhibits HMGB1, TLR4, and NF-κB activation and regulates the NLRP3 expression131,149
SinomenineInhibits the activation of NLRP3 inflammasomes, reduces the inflammatory cytokine levels
AllicinInhibits the NLRp3 signaling reduced pro-caspase-1, cleavage and IL-1β release133
MCC950Blocks the NLRP3/GSDMD signaling and ameliorates the liver injury and hepatocyte loss140

Emerging evidence suggests that the gut-liver axis plays a critical role in the pathogenesis of APAP-induced liver injury, and modulation of the gut microbiota and bile acid metabolism represents a novel therapeutic approach. Preclinical studies have shown that antibiotics, probiotics, and fecal microbiota transplantation can alter the gut microbiota composition and reduce liver injury in APAP overdose by modulating inflammatory responses and enhancing hepatocyte regeneration.155 Additionally, targeting bile acid metabolism, such as through bile acid sequestrants and farnesoid X receptor agonists, has shown promise in preclinical models by reducing bile acid accumulation and hepatotoxicity. Targeting inflammasome activation, inhibiting caspase-1, employing anti-inflammatory agents, and modulating the gut microbiota and bile acid metabolism represent promising avenues for developing novel treatments for this common and potentially life-threatening condition. Further research is needed to validate the efficacy and safety of these approaches in clinical settings and to identify optimal therapeutic regimens for patients with APAP overdose.

This review is inherently limited by its predominant focus on preclinical studies conducted in animal models or in vitro systems. While such models provide significant mechanistic insights into APAP-induced hepatotoxicity, including pathways related to oxidative stress, mitochondrial dysfunction, and inflammasome activation, they may not fully recapitulate the complexity of human physiology. Inter-species differences in drug metabolism, immune responses, and liver regeneration capacity can affect the translatability of findings, thereby restricting the extrapolation of preclinical data to clinical settings. Furthermore, the absence of strong data from clinical trials on novel therapeutic targets, such as inflammasome inhibitors or microbiome-modulating agents, limits the strength of conclusions regarding their efficacy and safety in humans. Although several drug molecules have shown hepatoprotective effects in experimental conditions, their clinical use remains inconclusive until validated in well-designed clinical trials. Another notable limitation is the scope of the populations discussed. This review does not comprehensively address APAP toxicity in specific vulnerable populations, such as neonates, pediatric patients, pregnant women, or the elderly, who may exhibit distinct pharmacokinetics, pharmacodynamics, and susceptibility to liver injury. These populations may require unique therapeutic approaches and risk stratification methods that were beyond the scope of the current review. Future studies should prioritize translational research efforts that bridge preclinical discoveries with clinical application, as well as investigate APAP-induced hepatotoxicity across diverse demographic and physiological subgroups.

Conclusions

Acetaminophen-induced liver injury remains a significant global public health concern due to its potential to cause acute liver failure and mortality in severe cases. The hepatotoxic effects of APAP are primarily mediated by its bioactivation to the reactive metabolite NAPQI via CYP2E1. Excessive accumulation of NAPQI depletes hepatic GSH stores, leading to oxidative stress, mitochondrial dysfunction, and covalent modification of cellular macromolecules. Multiple regulated and unregulated cell death pathways contribute to APAP-induced hepatotoxicity, including necrosis, apoptosis, necroptosis, and pyroptosis. Among these, sterile inflammation plays a pivotal role in amplifying liver injury. Inflammasome complexes, particularly NLRP3, have emerged as key regulators of this inflammatory response. Activation of the NLRP3 inflammasome leads to the cleavage of procaspase-1 into its active form, which in turn processes proinflammatory cytokines such as IL-1β and IL-18 and drives pyroptotic cell death via Gasdermin D activation. Despite the availability of NAC as the primary therapeutic agent for APAP overdose, treatment options remain limited, particularly when administered beyond the early therapeutic window. Consequently, current research has focused on identifying novel therapeutic strategies targeting the inflammatory and cell death pathways involved in APAP-induced hepatotoxicity. These include direct inhibition of inflammasome components, caspase-1 blockade, modulation of immune responses using anti-inflammatory agents, and targeted manipulation of gut microbiota and bile acid metabolism, which are increasingly recognized for their roles in liver homeostasis and injury response. Pharmacological inhibitors such as NLRP3 antagonists and caspase-1 inhibitors show promise in preclinical models but require further clinical validation. Emerging strategies targeting the gut-liver axis, such as microbiome modulation and bile acid receptor agonists, also offer new therapeutic avenues for APAP-induced liver injury. However, a deeper understanding of the molecular mechanisms leading to APAP-induced liver injury is crucial for developing effective, timely, and targeted therapies to reduce the global burden of APAP toxicity.

Declarations

Acknowledgement

The authors acknowledge DST-FIST for sponsoring the Department of Biotechnology, Central University of Tamil Nadu.

Funding

NS acknowledges the Science & Engineering Research Board and the University Grants Commission, Govt. of India, for a Junior Research Fellowship. CL acknowledges the Science & Engineering Research Board, Govt. of India, for Extra Mural Research Grant (EMR/2017/002793), the Indian Council of Medical Research, Govt. of India, for Extra Mural Research Grant (File Number: 36/14/2020/TOX/BMS), and UGC for Startup Research Grant.

Conflict of interest

The authors declare that they have no competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Authors’ contributions

Concept and design, reference collection, and writing of the manuscript (NS, LC). Both authors have approved the final version and publication of the manuscript.

References

  1. Lee WM. Acetaminophen (APAP) hepatotoxicity-Isn’t it time for APAP to go away?. J Hepatol 2017;67(6):1324-1331 View Article PubMed/NCBI
  2. Li X, Ni J, Chen L. Advances in the study of acetaminophen-induced liver injury. Front Pharmacol 2023;14:1239395 View Article PubMed/NCBI
  3. Heard KJ. Acetylcysteine for acetaminophen poisoning. N Engl J Med 2008;359(3):285-292 View Article PubMed/NCBI
  4. Yoon E, Babar A, Choudhary M, Kutner M, Pyrsopoulos N. Acetaminophen-Induced Hepatotoxicity: a Comprehensive Update. J Clin Transl Hepatol 2016;4(2):131-142 View Article PubMed/NCBI
  5. Chidiac AS, Buckley NA, Noghrehchi F, Cairns R. Paracetamol (acetaminophen) overdose and hepatotoxicity: mechanism, treatment, prevention measures, and estimates of burden of disease. Expert Opin Drug Metab Toxicol 2023;19(5):297-317 View Article PubMed/NCBI
  6. Green JL, Heard KJ, Reynolds KM, Albert D. Oral and Intravenous Acetylcysteine for Treatment of Acetaminophen Toxicity: A Systematic Review and Meta-analysis. West J Emerg Med 2013;14(3):218-226 View Article PubMed/NCBI
  7. Licata A, Minissale MG, Stankeviciute S, Sanabria-Cabrera J, Lucena MI, Andrade RJ, et al. N-Acetylcysteine for Preventing Acetaminophen-Induced Liver Injury: A Comprehensive Review. Front Pharmacol 2022;13:828565 View Article PubMed/NCBI
  8. Yang T, Wang H, Wang X, Li J, Jiang L. The Dual Role of Innate Immune Response in Acetaminophen-Induced Liver Injury. Biology (Basel) 2022;11(7):1057 View Article PubMed/NCBI
  9. Jaeschke H, McGill MR. Cytochrome P450-derived versus mitochondrial oxidant stress in acetaminophen hepatotoxicity. Toxicol Lett 2015;235(3):216-217 View Article PubMed/NCBI
  10. Chen W, Koenigs LL, Thompson SJ, Peter RM, Rettie AE, Trager WF, et al. Oxidation of acetaminophen to its toxic quinone imine and nontoxic catechol metabolites by baculovirus-expressed and purified human cytochromes P450 2E1 and 2A6. Chem Res Toxicol 1998;11(4):295-301 View Article PubMed/NCBI
  11. Leeming MG, Gamon LF, Wille U, Donald WA, O’Hair RA. What Are the Potential Sites of Protein Arylation by N-Acetyl-p-benzoquinone Imine (NAPQI)?. Chem Res Toxicol 2015;28(11):2224-2233 View Article PubMed/NCBI
  12. Luo G, Huang L, Zhang Z. The molecular mechanisms of acetaminophen-induced hepatotoxicity and its potential therapeutic targets. Exp Biol Med (Maywood) 2023;248(5):412-424 View Article PubMed/NCBI
  13. Kanno SI, Tomizawa A, Yomogida S, Hara A. Glutathione peroxidase 3 is a protective factor against acetaminophen-induced hepatotoxicity in vivo and in vitro. Int J Mol Med 2017;40(3):748-754 View Article PubMed/NCBI
  14. Andersson BS, Rundgren M, Nelson SD, Harder S. N-acetyl-p-benzoquinone imine-induced changes in the energy metabolism in hepatocytes. Chem Biol Interact 1990;75(2):201-211 View Article PubMed/NCBI
  15. Parmar DV, Ahmed G, Khandkar MA, Katyare SS. Mitochondrial ATPase: a target for paracetamol-induced hepatotoxicity. European journal of pharmacology 1995;293(3):225-229 View Article PubMed/NCBI
  16. Tirmenstein MA, Nelson SD. Acetaminophen-induced oxidation of protein thiols. Contribution of impaired thiol-metabolizing enzymes and the breakdown of adenine nucleotides. J Biol Chem 1990;265(6):3059-3065 PubMed/NCBI
  17. Ramachandran A, Jaeschke H. Acetaminophen hepatotoxicity: A mitochondrial perspective. Adv Pharmacol 2019;85:195-219 View Article PubMed/NCBI
  18. Hasan AA, Kalinina E, Tatarskiy V, Shtil A. The Thioredoxin System of Mammalian Cells and Its Modulators. Biomedicines 2022;10(7):1757 View Article PubMed/NCBI
  19. Hayakawa R, Hayakawa T, Takeda K, Ichijo H. Therapeutic targets in the ASK1-dependent stress signaling pathways. Proc Jpn Acad Ser B Phys Biol Sci 2012;88(8):434-453 View Article PubMed/NCBI
  20. Win S, Than TA, Zhang J, Oo C, Min RWM, Kaplowitz N. New insights into the role and mechanism of c-Jun-N-terminal kinase signaling in the pathobiology of liver diseases. Hepatology 2018;67(5):2013-2024 View Article PubMed/NCBI
  21. Heslop KA, Rovini A, Hunt EG, Fang D, Morris ME, Christie CF, et al. JNK activation and translocation to mitochondria mediates mitochondrial dysfunction and cell death induced by VDAC opening and sorafenib in hepatocarcinoma cells. Biochem Pharmacol 2020;171:113728 View Article PubMed/NCBI
  22. Latchoumycandane C, Goh CW, Ong MM, Boelsterli UA. Mitochondrial protection by the JNK inhibitor leflunomide rescues mice from acetaminophen-induced liver injury. Hepatology 2007;45(2):412-421 View Article PubMed/NCBI
  23. Zorov DB, Juhaszova M, Sollott SJ. Mitochondrial reactive oxygen species (ROS) and ROS-induced ROS release. Physiol Rev 2014;94(3):909-950 View Article PubMed/NCBI
  24. Bajt ML, Cover C, Lemasters JJ, Jaeschke H. Nuclear translocation of endonuclease G and apoptosis-inducing factor during acetaminophen-induced liver cell injury. Toxicol Sci 2006;94(1):217-225 View Article PubMed/NCBI
  25. Sevrioukova IF. Apoptosis-inducing factor: structure, function, and redox regulation. Antioxid Redox Signal 2011;14(12):2545-2579 View Article PubMed/NCBI
  26. Norberg E, Orrenius S, Zhivotovsky B. Mitochondrial regulation of cell death: processing of apoptosis-inducing factor (AIF). Biochem Biophys Res Commun 2010;396(1):95-100 View Article PubMed/NCBI
  27. Jaeschke H, Duan L, Akakpo JY, Farhood A, Ramachandran A. The role of apoptosis in acetaminophen hepatotoxicity. Food Chem Toxicol 2018;118:709-718 View Article PubMed/NCBI
  28. Huo Y, Yin S, Yan M, Win S, Aung Than T, Aghajan M, et al. Protective role of p53 in acetaminophen hepatotoxicity. Free Radic Biol Med 2017;106:111-117 View Article PubMed/NCBI
  29. Kruiswijk F, Labuschagne CF, Vousden KH. p53 in survival, death and metabolic health: a lifeguard with a licence to kill. Nat Rev Mol Cell Biol 2015;16(7):393-405 View Article PubMed/NCBI
  30. Copple IM, Goldring CE, Jenkins RE, Chia AJ, Randle LE, Hayes JD, et al. The hepatotoxic metabolite of acetaminophen directly activates the Keap1-Nrf2 cell defense system. Hepatology 2008;48(4):1292-1301 View Article PubMed/NCBI
  31. Ma Q. Role of nrf2 in oxidative stress and toxicity. Annu Rev Pharmacol Toxicol 2013;53:401-426 View Article PubMed/NCBI
  32. Velichkova M, Hasson T. Keap1 regulates the oxidation-sensitive shuttling of Nrf2 into and out of the nucleus via a Crm1-dependent nuclear export mechanism. Mol Cell Biol 2005;25(11):4501-4513 View Article PubMed/NCBI
  33. McJunkin B, Barwick KW, Little WC, Winfield JB. Fatal massive hepatic necrosis following acetaminophen overdose. JAMA 1976;236(16):1874-1875 PubMed/NCBI
  34. Jaeschke H, Ramachandran A. Acetaminophen Hepatotoxicity: Paradigm for Understanding Mechanisms of Drug-Induced Liver Injury. Annu Rev Pathol 2024;19:453-478 View Article PubMed/NCBI
  35. Gujral JS, Knight TR, Farhood A, Bajt ML, Jaeschke H. Mode of cell death after acetaminophen overdose in mice: apoptosis or oncotic necrosis?. Toxicol Sci 2002;67(2):322-328 View Article PubMed/NCBI
  36. Adams ML, Pierce RH, Vail ME, White CC, Tonge RP, Kavanagh TJ, et al. Enhanced acetaminophen hepatotoxicity in transgenic mice overexpressing BCL-2. Mol Pharmacol 2001;60(5):907-915 View Article PubMed/NCBI
  37. Jaeschke H, Cover C, Bajt ML. Role of caspases in acetaminophen-induced liver injury. Life Sci 2006;78(15):1670-1676 View Article PubMed/NCBI
  38. Belizario J, Vieira-Cordeiro L, Enns S. Necroptotic Cell Death Signaling and Execution Pathway: Lessons from Knockout Mice. Mediators Inflamm 2015;2015:128076 View Article PubMed/NCBI
  39. Lu Y, Zhang C, Chen YH, Wang H, Zhang ZH, Chen X, et al. Immature mice are more susceptible than adult mice to acetaminophen-induced acute liver injury. Sci Rep 2017;7:42736 View Article PubMed/NCBI
  40. Jaeschke H, Adelusi OB, Ramachandran A. Ferroptosis and Acetaminophen Hepatotoxicity: Are We Going Down Another Rabbit Hole?. Gene Expr 2021;20(3):169-178 View Article PubMed/NCBI
  41. Dara L, Johnson H, Suda J, Win S, Gaarde W, Han D, et al. Receptor interacting protein kinase 1 mediates murine acetaminophen toxicity independent of the necrosome and not through necroptosis. Hepatology 2015;62(6):1847-1857 View Article PubMed/NCBI
  42. Lin YH, Lin YC, Hou YT. Prospective Application of Tannic Acid in Acetaminophen (APAP)-Induced Acute Liver Failure. Int J Mol Sci 2023;25(1):317 View Article PubMed/NCBI
  43. Yamada N, Karasawa T, Takahashi M. Role of ferroptosis in acetaminophen-induced hepatotoxicity. Archives of toxicology 2020;94(5):1769-1770 View Article PubMed/NCBI
  44. Adelusi OB, Etemadi Y, Akakpo JY, Ramachandran A, Jaeschke H. Effect of ferroptosis inhibitors in a murine model of acetaminophen-induced liver injury. J Biochem Mol Toxicol 2024;38(8):e23791 View Article PubMed/NCBI
  45. Shi J, Gao W, Shao F. Pyroptosis: Gasdermin-Mediated Programmed Necrotic Cell Death. Trends Biochem Sci 2017;42(4):245-254 View Article PubMed/NCBI
  46. Yuan X, Chen P, Luan X, Yu C, Miao L, Zuo Y, et al. NLRP3 deficiency protects against acetaminophen-induced liver injury by inhibiting hepatocyte pyroptosis. Mol Med Rep 2024;29(4):61 View Article PubMed/NCBI
  47. Wang JC, Shi Q, Zhou Q, Zhang LL, Qiu YP, Lou DY, et al. Sapidolide A alleviates acetaminophen-induced acute liver injury by inhibiting NLRP3 inflammasome activation in macrophages. Acta Pharmacol Sin 2022;43(8):2016-2025 View Article PubMed/NCBI
  48. Jaeschke H, Umbaugh DS, Ramachandran A. Role of Pyroptosis in Acetaminophen-Induced Hepatotoxicity. Livers 2022;2(4):425-435 View Article PubMed/NCBI
  49. Williams CD, Farhood A, Jaeschke H. Role of caspase-1 and interleukin-1beta in acetaminophen-induced hepatic inflammation and liver injury. Toxicol Appl Pharmacol 2010;247(3):169-178 View Article PubMed/NCBI
  50. Woolbright BL, Nguyen NT, McGill MR, Sharpe MR, Curry SC, Jaeschke H. Generation of pro-and anti-inflammatory mediators after acetaminophen overdose in surviving and non-surviving patients. Toxicol Lett 2022;367:59-66 View Article PubMed/NCBI
  51. Shan Z, Ju C. Hepatic Macrophages in Liver Injury. Front Immunol 2020;11:322 View Article PubMed/NCBI
  52. Liaskou E, Wilson DV, Oo YH. Innate immune cells in liver inflammation. Mediators Inflamm 2012;2012:949157 View Article PubMed/NCBI
  53. De Gaetano A, Solodka K, Zanini G, Selleri V, Mattioli AV, Nasi M, et al. Molecular Mechanisms of mtDNA-Mediated Inflammation. Cells 2021;10(11):2898 View Article PubMed/NCBI
  54. Jaeschke H, Ramachandran A. Mechanisms and pathophysiological significance of sterile inflammation during acetaminophen hepatotoxicity. Food Chem Toxicol 2020;138:111240 View Article PubMed/NCBI
  55. Kawaratani H, Tsujimoto T, Douhara A, Takaya H, Moriya K, Namisaki T, et al. The effect of inflammatory cytokines in alcoholic liver disease. Mediators Inflamm 2013;2013:495156 View Article PubMed/NCBI
  56. Guo H, Chen S, Xie M, Zhou C, Zheng M. The complex roles of neutrophils in APAP-induced liver injury. Cell Prolif 2021;54(6):e13040 View Article PubMed/NCBI
  57. Imaeda AB, Watanabe A, Sohail MA, Mahmood S, Mohamadnejad M, Sutterwala FS, et al. Acetaminophen-induced hepatotoxicity in mice is dependent on Tlr9 and the Nalp3 inflammasome. J Clin Invest 2009;119(2):305-314 View Article PubMed/NCBI
  58. Yan M, Huo Y, Yin S, Hu H. Mechanisms of acetaminophen-induced liver injury and its implications for therapeutic interventions. Redox Biol 2018;17:274-283 View Article PubMed/NCBI
  59. Malik A, Kanneganti TD. Inflammasome activation and assembly at a glance. J Cell Sci 2017;130(23):3955-3963 View Article PubMed/NCBI
  60. Sharma M, de Alba E. Structure, Activation and Regulation of NLRP3 and AIM2 Inflammasomes. Int J Mol Sci 2021;22(2):872 View Article PubMed/NCBI
  61. Kodi T, Sankhe R, Gopinathan A, Nandakumar K, Kishore A. New Insights on NLRP3 Inflammasome: Mechanisms of Activation, Inhibition, and Epigenetic Regulation. J Neuroimmune Pharmacol 2024;19(1):7 View Article PubMed/NCBI
  62. de Zoete MR, Palm NW, Zhu S, Flavell RA. Inflammasomes. Cold Spring Harb Perspect Biol 2014;6(12):a016287 View Article PubMed/NCBI
  63. Brahadeeswaran S, Sivagurunathan N, Calivarathan L. Inflammasome Signaling in the Aging Brain and Age-Related Neurodegenerative Diseases. Mol Neurobiol 2022;59(4):2288-2304 View Article PubMed/NCBI
  64. Sivagurunathan N, Rahamathulla MP, Al-Dossary H, Calivarathan L. Emerging Role of Long Noncoding RNAs in Regulating Inflammasome-Mediated Neurodegeneration in Parkinson’s Disease. Molecular neurobiology 2024;61:4619-4632 View Article PubMed/NCBI
  65. Molla MD, Akalu Y, Geto Z, Dagnew B, Ayelign B, Shibabaw T. Role of Caspase-1 in the Pathogenesis of Inflammatory-Associated Chronic Noncommunicable Diseases. J Inflamm Res 2020;13:749-764 View Article PubMed/NCBI
  66. Kany S, Vollrath JT, Relja B. Cytokines in Inflammatory Disease. Int J Mol Sci 2019;20(23):6008 View Article PubMed/NCBI
  67. Li SJ, Liu AB, Yu YY, Ma JH. The role and mechanism of pyroptosis and potential therapeutic targets in non-alcoholic fatty liver disease (NAFLD). Front Cell Dev Biol 2024;12:1407738 View Article PubMed/NCBI
  68. Sun J, Chen C, Wang J. Artesunate Inhibits Lipid Accumulation and Inflammation by Regulating the NLRP3 Inflammasome in Nonalcoholic Fatty Liver Disease. Discov Med 2024;36(181):385-392 View Article PubMed/NCBI
  69. Mitsuyoshi H, Yasui K, Hara T, Taketani H, Ishiba H, Okajima A, et al. Hepatic nucleotide binding oligomerization domain-like receptors pyrin domain-containing 3 inflammasomes are associated with the histologic severity of non-alcoholic fatty liver disease. Hepatol Res 2017;47(13):1459-1468 View Article PubMed/NCBI
  70. Petrasek J, Bala S, Csak T, Lippai D, Kodys K, Menashy V, et al. IL-1 receptor antagonist ameliorates inflammasome-dependent alcoholic steatohepatitis in mice. J Clin Invest 2012;122(10):3476-3489 View Article PubMed/NCBI
  71. Zou J, Yang R, Feng R, Liu J, Wan JB. Ginsenoside Rk2, a dehydroprotopanaxadiol saponin, alleviates alcoholic liver disease via regulating NLRP3 and NLRP6 inflammasome signaling pathways in mice. J Pharm Anal 2023;13(9):999-1012 View Article PubMed/NCBI
  72. Wei B, Li H, Cheng M, Yang Y, Liu B, Tian Y, et al. NLRP3 Inflammasome Activation Mediates Hepatitis E Virus-Induced Neuroinflammation. J Viral Hepat 2024;31(11):729-738 View Article PubMed/NCBI
  73. Li Z, Liu T, Feng Y, Tong Y, Jia Y, Wang C, et al. PPARgamma Alleviates Sepsis-Induced Liver Injury by Inhibiting Hepatocyte Pyroptosis via Inhibition of the ROS/TXNIP/NLRP3 Signaling Pathway. Oxid Med Cell Longev 2022;2022:1269747 View Article PubMed/NCBI
  74. Menini S, Iacobini C, Vitale M, Pugliese G. The Inflammasome in Chronic Complications of Diabetes and Related Metabolic Disorders. Cells 2020;9(8):1812 View Article PubMed/NCBI
  75. Huebener P, Hernandez C, Schwabe RF. HMGB1 and injury amplification. Oncotarget 2015;6(27):23048-23049 View Article PubMed/NCBI
  76. McGill MR, Sharpe MR, Williams CD, Taha M, Curry SC, Jaeschke H. The mechanism underlying acetaminophen-induced hepatotoxicity in humans and mice involves mitochondrial damage and nuclear DNA fragmentation. J Clin Invest 2012;122(4):1574-1583 View Article PubMed/NCBI
  77. Kono H, Chen CJ, Ontiveros F, Rock KL. Uric acid promotes an acute inflammatory response to sterile cell death in mice. J Clin Invest 2010;120(6):1939-1949 View Article PubMed/NCBI
  78. Martin-Murphy BV, Holt MP, Ju C. The role of damage associated molecular pattern molecules in acetaminophen-induced liver injury in mice. Toxicol Lett 2010;192(3):387-394 View Article PubMed/NCBI
  79. Blevins HM, Xu Y, Biby S, Zhang S. The NLRP3 Inflammasome Pathway: A Review of Mechanisms and Inhibitors for the Treatment of Inflammatory Diseases. Front Aging Neurosci 2022;14:879021 View Article PubMed/NCBI
  80. Kelley N, Jeltema D, Duan Y, He Y. The NLRP3 Inflammasome: An Overview of Mechanisms of Activation and Regulation. Int J Mol Sci 2019;20(13):3328 View Article PubMed/NCBI
  81. Munoz-Planillo R, Kuffa P, Martinez-Colon G, Smith BL, Rajendiran TM, Nunez G. K(+) efflux is the common trigger of NLRP3 inflammasome activation by bacterial toxins and particulate matter. Immunity 2013;38(6):1142-1153 View Article PubMed/NCBI
  82. Liu Y, Guo ZW, Li J, Li AH, Huo TG. Insight into the regulation of NLRP3 inflammasome activation by mitochondria in liver injury and the protective role of natural products. Biomed Pharmacother 2022;156:113968 View Article PubMed/NCBI
  83. Grossini E, Garhwal DP, Calamita G, Romito R, Rigamonti C, Minisini R, et al. Exposure to Plasma From Non-alcoholic Fatty Liver Disease Patients Affects Hepatocyte Viability, Generates Mitochondrial Dysfunction, and Modulates Pathways Involved in Fat Accumulation and Inflammation. Front Med (Lausanne) 2021;8:693997 View Article PubMed/NCBI
  84. Huang YJ, Chen P, Lee CY, Yang SY, Lin MT, Lee HS, et al. Protection against acetaminophen-induced acute liver failure by omentum adipose tissue derived stem cells through the mediation of Nrf2 and cytochrome P450 expression. J Biomed Sci 2016;23:5 View Article PubMed/NCBI
  85. Xun T, Zhang M, Wei S, Zhao C, Lin Z, Feng H, et al. CYP2E1 mediated advanced oxidation protein products exacerbate acetaminophen induced drug-derived liver injury in vitro and in vivo. Eur J Pharm Sci 2024;200:106829 View Article PubMed/NCBI
  86. Grazioli S, Pugin J. Mitochondrial Damage-Associated Molecular Patterns: From Inflammatory Signaling to Human Diseases. Front Immunol 2018;9:832 View Article PubMed/NCBI
  87. Shimada K, Crother TR, Karlin J, Dagvadorj J, Chiba N, Chen S, et al. Oxidized mitochondrial DNA activates the NLRP3 inflammasome during apoptosis. Immunity 2012;36(3):401-414 View Article PubMed/NCBI
  88. Nakahira K, Haspel JA, Rathinam VA, Lee SJ, Dolinay T, Lam HC, et al. Autophagy proteins regulate innate immune responses by inhibiting the release of mitochondrial DNA mediated by the NALP3 inflammasome. Nat Immunol 2011;12(3):222-230 View Article PubMed/NCBI
  89. Krysko DV, Agostinis P, Krysko O, Garg AD, Bachert C, Lambrecht BN, et al. Emerging role of damage-associated molecular patterns derived from mitochondria in inflammation. Trends Immunol 2011;32(4):157-164 View Article PubMed/NCBI
  90. Liu J, Wang T, He K, Xu M, Gong JP. Cardiolipin inhibitor ameliorates the non-alcoholic steatohepatitis through suppressing NLRP3 inflammasome activation. Eur Rev Med Pharmacol Sci 2019;23(18):8158-8167 View Article PubMed/NCBI
  91. Yabal M, Calleja DJ, Simpson DS, Lawlor KE. Stressing out the mitochondria: Mechanistic insights into NLRP3 inflammasome activation. J Leukoc Biol 2019;105(2):377-399 View Article PubMed/NCBI
  92. Salas VM, Corcoran GB. Calcium-dependent DNA damage and adenosine 3′,5′-cyclic monophosphate-independent glycogen phosphorylase activation in an in vitro model of acetaminophen-induced liver injury. Hepatology 1997;25(6):1432-1438 View Article PubMed/NCBI
  93. Lee GS, Subramanian N, Kim AI, Aksentijevich I, Goldbach-Mansky R, Sacks DB, et al. The calcium-sensing receptor regulates the NLRP3 inflammasome through Ca2+ and cAMP. Nature 2012;492(7427):123-127 View Article PubMed/NCBI
  94. Ono H, Ohta R, Kawasaki Y, Niwa A, Takada H, Nakahata T, et al. Lysosomal membrane permeabilization causes secretion of IL-1beta in human vascular smooth muscle cells. Inflamm Res 2018;67(10):879-889 View Article PubMed/NCBI
  95. Alu A, Han X, Ma X, Wu M, Wei Y, Wei X. The role of lysosome in regulated necrosis. Acta Pharm Sin B 2020;10(10):1880-1903 View Article PubMed/NCBI
  96. Scaffidi P, Misteli T, Bianchi ME. Release of chromatin protein HMGB1 by necrotic cells triggers inflammation. Nature 2002;418(6894):191-195 View Article PubMed/NCBI
  97. Li W, Deng M, Loughran PA, Yang M, Lin M, Yang C, et al. LPS Induces Active HMGB1 Release From Hepatocytes Into Exosomes Through the Coordinated Activities of TLR4 and Caspase-11/GSDMD Signaling. Front Immunol 2020;11:229 View Article PubMed/NCBI
  98. Volchuk A, Ye A, Chi L, Steinberg BE, Goldenberg NM. Indirect regulation of HMGB1 release by gasdermin D. Nat Commun 2020;11(1):4561 View Article PubMed/NCBI
  99. Sayaf K, Battistella S, Russo FP. NLRP3 Inflammasome in Acute and Chronic Liver Diseases. Int J Mol Sci 2024;25(8):4537 View Article PubMed/NCBI
  100. Le Dare B, Ferron PJ, Gicquel T. The Purinergic P2X7 Receptor-NLRP3 Inflammasome Pathway: A New Target in Alcoholic Liver Disease?. Int J Mol Sci 2021;22(4):2139 View Article PubMed/NCBI
  101. Liu T, Zhang L, Joo D, Sun SC. NF-kappaB signaling in inflammation. Sig Transduct Target Ther 2017;2:17023 View Article PubMed/NCBI
  102. Cipriano A, Viviano M, Feoli A, Milite C, Sarno G, Castellano S, et al. NADPH Oxidases: From Molecular Mechanisms to Current Inhibitors. J Med Chem 2023;66(17):11632-11655 View Article PubMed/NCBI
  103. Tschopp J, Schroder K. NLRP3 inflammasome activation: The convergence of multiple signalling pathways on ROS production?. Nat Rev Immunol 2010;10(3):210-215 View Article PubMed/NCBI
  104. Houshmandfar S, Saeedi-Boroujeni A, Rashno M, Khodadadi A, Mahmoudian-Sani MR. miRNA-223 as a regulator of inflammation and NLRP3 inflammasome, the main fragments in the puzzle of immunopathogenesis of different inflammatory diseases and COVID-19. Naunyn Schmiedebergs Arch Pharmacol 2021;394(11):2187-2195 View Article PubMed/NCBI
  105. Wan L, Yuan X, Liu M, Xue B. miRNA-223-3p regulates NLRP3 to promote apoptosis and inhibit proliferation of hep3B cells. Exp Ther Med 2018;15(3):2429-2435 View Article PubMed/NCBI
  106. Qiao Y, Wang P, Qi J, Zhang L, Gao C. TLR-induced NF-kappaB activation regulates NLRP3 expression in murine macrophages. FEBS Lett 2012;586(7):1022-1026 View Article PubMed/NCBI
  107. Sutterwala FS, Haasken S, Cassel SL. Mechanism of NLRP3 inflammasome activation. Ann N Y Acad Sci 2014;1319(1):82-95 View Article PubMed/NCBI
  108. Zhu L, Wang Z, Sun X, Yu J, Li T, Zhao H, et al. STAT3/Mitophagy Axis Coordinates Macrophage NLRP3 Inflammasome Activation and Inflammatory Bone Loss. J Bone Miner Res 2023;38(2):335-353 View Article PubMed/NCBI
  109. Franchi L, Eigenbrod T, Munoz-Planillo R, Nunez G. The inflammasome: a caspase-1-activation platform that regulates immune responses and disease pathogenesis. Nat Immunol 2009;10(3):241-247 View Article PubMed/NCBI
  110. Fernandes-Alnemri T, Wu J, Yu JW, Datta P, Miller B, Jankowski W, et al. The pyroptosome: a supramolecular assembly of ASC dimers mediating inflammatory cell death via caspase-1 activation. Cell Death Differ 2007;14(9):1590-1604 View Article PubMed/NCBI
  111. Martinon F, Burns K, Tschopp J. The inflammasome: a molecular platform triggering activation of inflammatory caspases and processing of proIL-beta. Mol Cell 2002;10(2):417-426 View Article PubMed/NCBI
  112. Dinarello CA. Immunological and inflammatory functions of the interleukin-1 family. Annu Rev Immunol 2009;27:519-550 View Article PubMed/NCBI
  113. Shi J, Zhao Y, Wang K, Shi X, Wang Y, Huang H, et al. Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nature 2015;526(7575):660-665 View Article PubMed/NCBI
  114. Kayagaki N, Stowe IB, Lee BL, O’Rourke K, Anderson K, Warming S, et al. Caspase-11 cleaves gasdermin D for non-canonical inflammasome signalling. Nature 2015;526(7575):666-671 View Article PubMed/NCBI
  115. He WT, Wan H, Hu L, Chen P, Wang X, Huang Z, et al. Gasdermin D is an executor of pyroptosis and required for interleukin-1beta secretion. Cell Res 2015;25(12):1285-1298 View Article PubMed/NCBI
  116. Jorgensen I, Lopez JP, Laufer SA, Miao EA. IL-1beta, IL-18, and eicosanoids promote neutrophil recruitment to pore-induced intracellular traps following pyroptosis. Eur J Immunol 2016;46(12):2761-2766 View Article PubMed/NCBI
  117. Chen R, Kang R, Tang D. The mechanism of HMGB1 secretion and release. Exp Mol Med 2022;54(2):91-102 View Article PubMed/NCBI
  118. Willingham SB, Allen IC, Bergstralh DT, Brickey WJ, Huang MT, Taxman DJ, et al. NLRP3 (NALP3, Cryopyrin) facilitates in vivo caspase-1 activation, necrosis, and HMGB1 release via inflammasome-dependent and -independent pathways. J Immunol 2009;183(3):2008-2015 View Article PubMed/NCBI
  119. Lawson JA, Farhood A, Hopper RD, Bajt ML, Jaeschke H. The hepatic inflammatory response after acetaminophen overdose: role of neutrophils. Toxicol Sci 2000;54(2):509-516 View Article PubMed/NCBI
  120. Koyama Y, Brenner DA. Liver inflammation and fibrosis. J Clin Invest 2017;127(1):55-64 View Article PubMed/NCBI
  121. Pradere JP, Kluwe J, De Minicis S, Jiao JJ, Gwak GY, Dapito DH, et al. Hepatic macrophages but not dendritic cells contribute to liver fibrosis by promoting the survival of activated hepatic stellate cells in mice. Hepatology 2013;58(4):1461-1473 View Article PubMed/NCBI
  122. Jaffer U, Wade RG, Gourlay T. Cytokines in the systemic inflammatory response syndrome: a review. HSR Proc Intensive Care Cardiovasc Anesth 2010;2(3):161-175 PubMed/NCBI
  123. Li J, Mbow ML, Sun L, Li L, Yang G, Griswold DE, et al. Induction of dendritic cell maturation by IL-18. Cell Immunol 2004;227(2):103-108 View Article PubMed/NCBI
  124. Van Den Eeckhout B, Tavernier J, Gerlo S. Interleukin-1 as Innate Mediator of T Cell Immunity. Front Immunol 2020;11:621931 View Article PubMed/NCBI
  125. Li L, Shan S, Kang K, Zhang C, Kou R, Song F. The cross-talk of NLRP3 inflammasome activation and necroptotic hepatocyte death in acetaminophen-induced mice acute liver injury. Hum Exp Toxicol 2021;40(4):673-684 View Article PubMed/NCBI
  126. Wree A, Eguchi A, McGeough MD, Pena CA, Johnson CD, Canbay A, et al. NLRP3 inflammasome activation results in hepatocyte pyroptosis, liver inflammation, and fibrosis in mice. Hepatology 2014;59(3):898-910 View Article PubMed/NCBI
  127. Shen XL, Guo YN, Lu MH, Ding KN, Liang SS, Mou RW, et al. Acetaminophen-induced hepatotoxicity predominantly via inhibiting Nrf2 antioxidative pathway and activating TLR4-NF-kappaB-MAPK inflammatory response in mice. Ecotoxicol Environ Saf 2023;252:114590 View Article PubMed/NCBI
  128. Jiang L, Ke M, Yue S, Xiao W, Yan Y, Deng X, et al. Blockade of Notch signaling promotes acetaminophen-induced liver injury. Immunol Res 2017;65(3):739-749 View Article PubMed/NCBI
  129. Li ZC, Xu FF, Fu JT, Ouyang SX, Cao Q, Yan YY, et al. Sting mutation attenuates acetaminophen-induced acute liver injury by limiting NLRP3 activation. Int Immunopharmacol 2023;125(Pt A):111133 View Article PubMed/NCBI
  130. Kim EH, Park PH. Globular adiponectin protects rat hepatocytes against acetaminophen-induced cell death via modulation of the inflammasome activation and ER stress: Critical role of autophagy induction. Biochem Pharmacol 2018;154:278-292 View Article PubMed/NCBI
  131. Du YC, Lai L, Zhang H, Zhong FR, Cheng HL, Qian BL, et al. Kaempferol from Penthorum chinense Pursh suppresses HMGB1/TLR4/NF-kappaB signaling and NLRP3 inflammasome activation in acetaminophen-induced hepatotoxicity. Food Funct 2020;11(9):7925-7934 View Article PubMed/NCBI
  132. Ramalingam V. NLRP3 inhibitors: Unleashing their therapeutic potential against inflammatory diseases. Biochem Pharmacol 2023;218:115915 View Article PubMed/NCBI
  133. Samra YA, Hamed MF, El-Sheakh AR. Hepatoprotective effect of allicin against acetaminophen-induced liver injury: Role of inflammasome pathway, apoptosis, and liver regeneration. J Biochem Mol Toxicol 2020;34(5):e22470 View Article PubMed/NCBI
  134. Vivoli E, Cappon A, Milani S, Piombanti B, Provenzano A, Novo E, et al. NLRP3 inflammasome as a target of berberine in experimental murine liver injury: interference with P2X7 signalling. Clin Sci (Lond) 2016;130(20):1793-1806 View Article PubMed/NCBI
  135. Yu D, Li J, Wang Y, Guo D, Zhang X, Chen M, et al. Oridonin ameliorates acetaminophen-induced acute liver injury through ATF4/PGC-1alpha pathway. Drug Dev Res 2023;84(2):211-225 View Article PubMed/NCBI
  136. Hu C, Li M, Chen Y, Cheng W, Wang H, Zhou Y, et al. AIM2 regulates autophagy to mitigate oxidative stress in aged mice with acute liver injury. Cell Death Discov 2024;10(1):107 View Article PubMed/NCBI
  137. Williams CD, Antoine DJ, Shaw PJ, Benson C, Farhood A, Williams DP, et al. Role of the Nalp3 inflammasome in acetaminophen-induced sterile inflammation and liver injury. Toxicol Appl Pharmacol 2011;252(3):289-297 View Article PubMed/NCBI
  138. Ioannou GN, Horn CL, Kothari V, Yeh MM, Shyu I, Lee SP, et al. Genetic deletion or pharmacologic inhibition of the Nlrp3 inflammasome did not ameliorate experimental NASH. J Lipid Res 2023;64(2):100330 View Article PubMed/NCBI
  139. Zhang X, Xu A, Lv J, Zhang Q, Ran Y, Wei C, et al. Development of small molecule inhibitors targeting NLRP3 inflammasome pathway for inflammatory diseases. Eur J Med Chem 2020;185:111822 View Article PubMed/NCBI
  140. Yan W, Shen Y, Huang J, Lu L, Zhang Q. MCC950 Ameliorates Acute Liver Injury Through Modulating Macrophage Polarization and Myeloid-Derived Suppressor Cells Function. Front Med (Lausanne) 2021;8:752223 View Article PubMed/NCBI
  141. Yuan Y, Zhang J, Li H, Yuan F, Cui Q, Wu D, et al. Scopoletin alleviates acetaminophen-induced hepatotoxicity through modulation of NLRP3 inflammasome activation and Nrf2/HMGB1/TLR4/NF-kappaB signaling pathway. Int Immunopharmacol 2025;148:114132 View Article PubMed/NCBI
  142. Coelho AM, Queiroz IF, Perucci LO, Menezes TP, Lima WG, Talvani A, et al. Piperine as an Herbal Alternative for the Prevention of Drug-Induced Liver Damage Caused by Paracetamol. Pharmaceuticals (Basel) 2024;17(11):1477 View Article PubMed/NCBI
  143. Chen H, Wang S, Chen Q, Yu W, Nie H, Liu L, et al. Aloperine Ameliorates Acetaminophen-Induced Acute Liver Injury through HMGB1/TLR4/NF-kappaB and NLRP3/Inflammasome Pathway. Mediators Inflamm 2024;2024:3938136 View Article PubMed/NCBI
  144. Gao Z, Zhan H, Zong W, Sun M, Linghu L, Wang G, et al. Salidroside alleviates acetaminophen-induced hepatotoxicity via Sirt1-mediated activation of Akt/Nrf2 pathway and suppression of NF-kappaB/NLRP3 inflammasome axis. Life Sci 2023;327:121793 View Article PubMed/NCBI
  145. Zhao R, Zhang Q, Liu W, Lin Y, He Y, Chang D, et al. Pien Tze Huang attenuated acetaminophen-induced liver injury by autophagy mediated-NLRP3 inflammasome inhibition. J Ethnopharmacol 2023;311:116285 View Article PubMed/NCBI
  146. Yao Y, Li R, Liu D, Long L, He N. Rosmarinic acid alleviates acetaminophen-induced hepatotoxicity by targeting Nrf2 and NEK7-NLRP3 signaling pathway. Ecotoxicol Environ Saf 2022;241:113773 View Article PubMed/NCBI
  147. Shen P, Han L, Chen G, Cheng Z, Liu Q. Emodin Attenuates Acetaminophen-Induced Hepatotoxicity via the cGAS-STING Pathway. Inflammation 2022;45(1):74-87 View Article PubMed/NCBI
  148. Wang Y, Zhao Y, Wang Z, Sun R, Zou B, Li R, et al. Peroxiredoxin 3 Inhibits Acetaminophen-Induced Liver Pyroptosis Through the Regulation of Mitochondrial ROS. Front Immunol 2021;12:652782 View Article PubMed/NCBI
  149. Li H, Weng Q, Gong S, Zhang W, Wang J, Huang Y, et al. Kaempferol prevents acetaminophen-induced liver injury by suppressing hepatocyte ferroptosis via Nrf2 pathway activation. Food Funct 2023;14(4):1884-1896 View Article PubMed/NCBI
  150. Litwiniuk A, Baranowska-Bik A, Domanska A, Kalisz M, Bik W. Contribution of Mitochondrial Dysfunction Combined with NLRP3 Inflammasome Activation in Selected Neurodegenerative Diseases. Pharmaceuticals (Basel) 2021;14(12):1221 View Article PubMed/NCBI
  151. Yang M, Zhao L. The Selective NLRP3-Inflammasome Inhibitor CY-09 Ameliorates Kidney Injury in Diabetic Nephropathy by Inhibiting NLRP3- inflammasome Activation. Curr Med Chem 2023;30(28):3261-3270 View Article PubMed/NCBI
  152. Qu J, Yuan Z, Wang G, Wang X, Li K. The selective NLRP3 inflammasome inhibitor MCC950 alleviates cholestatic liver injury and fibrosis in mice. Int Immunopharmacol 2019;70:147-155 View Article PubMed/NCBI
  153. Chen H, Wang Y, Jiao FZ, Yang F, Li X, Wang LW. Sinomenine Attenuates Acetaminophen-Induced Acute Liver Injury by Decreasing Oxidative Stress and Inflammatory Response via Regulating TGF-beta/Smad Pathway in vitro and in vivo. Drug Des Devel Ther 2020;14:2393-2403 View Article PubMed/NCBI
  154. MacKenzie SH, Schipper JL, Clark AC. The potential for caspases in drug discovery. Curr Opin Drug Discov Devel 2010;13(5):568-576 PubMed/NCBI
  155. Jiang L, Schnabl B. Gut Microbiota in Liver Disease: What Do We Know and What Do We Not Know?. Physiology (Bethesda) 2020;35(4):261-274 View Article PubMed/NCBI
  156. Bindu S, Mazumder S, Bandyopadhyay U. Non-steroidal anti-inflammatory drugs (NSAIDs) and organ damage: A current perspective. Biochem Pharmacol 2020;180:114147 View Article PubMed/NCBI
  157. Kaya Tektemur N, Erdem Guzel E, Gul M, Tektemur A, Ozcan Yildirim S, Kavak Balgetir M, et al. The combination of N-acetylcysteine and cyclosporin A reduces acetaminophen-induced hepatotoxicity in mice. Ultrastruct Pathol 2021;45(1):19-27 View Article PubMed/NCBI

About this Article

Cite this article
Sivagurunathan N, Calivarathan L. Inflammasome Activation as a Key Driver of Acetaminophen-induced Hepatotoxicity: Mechanisms and Emerging Therapeutics. Gene Expr. Published online: Jul 21, 2025. doi: 10.14218/GE.2025.00001.
Copy        Export to RIS        Export to EndNote
Article History
Received Revised Accepted Published
January 1, 2025 May 10, 2025 May 30, 2025 July 21, 2025
DOI http://dx.doi.org/10.14218/GE.2025.00001
  • Gene Expression
  • eISSN 1555-3884
Back to Top

Inflammasome Activation as a Key Driver of Acetaminophen-induced Hepatotoxicity: Mechanisms and Emerging Therapeutics

Narmadhaa Sivagurunathan, Latchoumycandane Calivarathan
  • Reset Zoom
  • Download TIFF