Back to Journals » Journal of Inflammation Research » Volume 18

PANoptosis in Sepsis: A Central Role and Emerging Therapeutic Target

Authors Wu Q, Qi S , Kang Z, Bai X, Li Z , Cheng J, Dong X

Received 20 December 2024

Accepted for publication 4 May 2025

Published 13 May 2025 Volume 2025:18 Pages 6245—6261

DOI https://doi.org/10.2147/JIR.S513367

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 2

Editor who approved publication: Dr Adam Bachstetter



Qiqi Wu,* Siyuan Qi,* Zhaofeng Kang, Xiangjun Bai, Zhanfei Li, Jing Cheng, Xijie Dong

Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China

*These authors contributed equally to this work

Correspondence: Jing Cheng, Email [email protected] Xijie Dong, Email [email protected]

Abstract: The pathogenesis of sepsis is intricately linked to regulated cell death. As a novel form of regulated cell death, PANoptosis plays a critical role in driving the inflammatory response, impairing immune cell function, and contributing to multi-organ dysfunction in sepsis. This review explores the molecular mechanisms underlying PANoptosis and its involvement in sepsis. By activating multiple pathways, PANoptosis promotes the release of inflammatory cytokines, triggering a cytokine storm that disrupts immune cell homeostasis and exacerbates organ damage. Emerging therapeutic strategies targeting PANoptosis, including chemotherapeutic agents and herbal remedies, are showing potential for clinical application. The concept of targeting PANoptosis offers a promising avenue for developing innovative treatments for sepsis.

Keywords: PANoptosis, regulated cell death, sepsis, therapeutic target

Introduction

Sepsis is a life-threatening condition characterized by organ dysfunction caused by a dysregulated host response to infection. The 2016 Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3) clarified this concept, highlighting the critical role of infection as a trigger and the importance of the host’s dysregulated response.1 Sepsis remains a major global health challenge, with millions of cases reported annually. It is estimated that approximately 31.5 million cases of sepsis occur worldwide each year, leading to around 5.3 million deaths. Even survivors often experience long-term health complications.2 The incidence and mortality rates vary by region. In low-income countries, sepsis poses a heavier burden due to the high prevalence of infectious diseases such as malaria, Human Immunodeficiency Virus (HIV), and dengue fever. In high-income countries, despite advanced healthcare systems, sepsis remains a leading cause of mortality.2,3

Sepsis affects multiple organ systems, leading to a wide range of clinical manifestations. Patients with sepsis often experience arterial hypotension, which results from a combination of factors such as reduced blood volume, vascular tone loss, and myocardial suppression.4 Hypotension reduces cardiac output, leading to inadequate tissue perfusion and impaired organ function. Sepsis-induced myocardial injury manifests as decreased myocardial contractility and diastolic dysfunction.5,6 Increased apoptosis and necroptosis of cardiomyocytes are key contributors to cardiac dysfunction, likely driven by inflammatory cytokines, oxidative stress, and mitochondrial dysfunction.7,8 Respiratory abnormalities such as tachypnea and hypoxemia are also common in sepsis. Pulmonary inflammation damages the alveolar-capillary membrane, disrupting gas exchange and causing respiratory distress. Severe cases may develop acute respiratory distress syndrome (ARDS), severely impairing lung ventilation and gas exchange, often requiring mechanical ventilation to sustain life.9,10 Sepsis frequently causes kidney damage, presenting as oliguria or anuria, along with elevated serum creatinine and blood urea nitrogen levels. Acute kidney injury (AKI) is a common complication, driven by inflammation, oxidative stress, apoptosis, and necrosis. AKI not only impairs renal excretory function but also exacerbates systemic inflammation and multi-organ dysfunction, significantly increasing mortality.11–13 Beyond these, sepsis impacts the nervous, hematologic, and hepatic systems, contributing to its complex and multifaceted clinical presentation.14–17 In sepsis, various forms of regulated cell death (RCD)—including apoptosis, necroptosis, and pyroptosis—play critical roles in disease progression.18 PANoptosis, a recently proposed concept that integrates apoptosis, necroptosis, and pyroptosis, has also been found to be closely associated with sepsis. Apoptosis, a classical RCD pathway, is characterized by membrane blebbing, DNA fragmentation, and a lack of inflammatory response.19 Pyroptosis, on the other hand, involves inflammasome activation and membrane pore formation, leading to the release of inflammatory cytokines.20 Necroptosis features molecules like Receptor-Interacting Protein Kinase 1 (RIPK1), Receptor-Interacting Protein Kinase 3 (RIPK3), and Mixed Lineage Kinase Domain-Like Protein (MLKL), resulting in membrane rupture and inflammatory responses.21 In contrast, PANoptosis represents a more intricate RCD pathway. It integrates key molecules from these pathways, such as inflammasome sensors, Caspase-8, RIPK1/3, and GSDMD, forming a multifunctional molecular platform known as the PANoptosome. This platform orchestrates the cell death process by regulating the interactions and cross-talk among these pathways.22 The molecular mechanisms of PANoptosis are significantly more complex, involving not only the activation of individual pathways but also their dynamic interplay. This interplay allows for a more precise and adaptive regulation of cell death, highlighting the unique role of PANoptosis in cellular processes.

Sepsis and Regulated Cell Death

RCD plays a critical role in the progression of sepsis. During the pathophysiology of sepsis, apoptosis is prevalent in various cell types, including lymphocytes, monocytes, neutrophils, endothelial cells, and epithelial cells.23–26 Factors such as pathogen infection, inflammatory cytokine release, oxidative stress, and mitochondrial damage can activate apoptotic pathways. Apoptosis has a dual role in sepsis: moderate levels help eliminate infected and damaged cells, reducing pathogen replication and alleviating inflammation.27 However, excessive apoptosis depletes cell populations, impairs immune function, increases infection risk, exacerbates tissue damage, and contributes to multi-organ dysfunction.25,28 Necroptosis, a regulated form of cell death with necrotic characteristics, combines the programmed nature of apoptosis with necrosis-like morphology.21,27 In sepsis, necroptosis pathways are activated, leading to cell death. Studies suggest that inhibiting necroptosis can reduce inflammation, limit cell death, improve organ function, and lower mortality, making it a potential therapeutic target.29 Pyroptosis, an inflammatory form of RCD, is mediated by inflammatory caspases (caspase-1, −4, −5, and −11).27 In sepsis, pathogen infections induce inflammasome assembly, activating these caspases to cleave substrates such as Gasdermin D (GSDMD) and Gasdermin D (GSDME). This results in cell swelling, rupture, and the release of inflammatory cytokines and intracellular contents, triggering inflammatory responses. While moderate pyroptosis helps control infection and clear pathogens, excessive pyroptosis leads to uncontrolled inflammation, worsening tissue damage.30,31 Pyroptosis is also closely linked to sepsis-associated organ dysfunction, including injuries to the kidneys, liver, and lungs.32

Sepsis involves additional forms of cell death, such as autophagy and ferroptosis, which also play significant roles in its progression.30,33 Research has shown that cuproptosis is associated with the onset of septic cardiomyopathy and immune infiltration.34 The pathological process of sepsis promotes cuproptosis in cardiomyocytes, resulting in cardiac toxicity.35 Cuproptosis also plays a critical role in the human immune response, with several key genes closely related to the prognosis of sepsis patients.36,37 Immune cells such as macrophages, T cells, and B cells undergo ferroptosis, which leads to their reduced number and function. Ferroptotic cells can be recognized by immune cells, triggering a cascade of inflammatory or specific immune responses.38 Ferroptosis also contributes to septic myocardial injury. Inhibiting ferroptosis can significantly alleviate LPS-induced cardiac damage and inflammation in mice.39 It has been confirmed that ferroptosis plays a key role in LPS-induced acute lung injury.40 Ferroptosis is also critically involved in diseases such as acute kidney injury and sepsis-associated encephalopathy (SAE).41 Autophagy, closely linked to inflammation and immunity, provides protective effects in sepsis by negatively regulating abnormal macrophage activation, modulating macrophage polarization, reducing inflammasome activation, and limiting the release of inflammatory cytokines.42,43 Autophagy supports antigen presentation and T cell homeostasis, influencing T cell function and polarization.44 However, excessive autophagy may lead to autophagic death of macrophages, exacerbating the inflammatory response.42 Some studies have shown that autophagy also contributes to mitochondrial damage induced by sepsis, potentially resulting in harmful effects on the body.45 Excessive autophagy increases regulated cell death.46 Enhanced autophagy in pulmonary endothelial cells can lead to increased vascular permeability, worsening septic lung injury.47 Understanding the mechanisms of RCD in sepsis is essential for identifying novel therapeutic targets.

PANoptosis and the Maintenance of Homeostasis

PANoptosis is a recently identified form of RCD, first proposed by Malireddi et al in 2019.48 This concept emerged from extensive research into cell death mechanisms, particularly the crosstalk between apoptosis, pyroptosis, and necroptosis (Figure 1). These three RCD pathways are not entirely independent but form an intricate network of interactions and regulatory mechanisms.49 The hallmark of PANoptosis is its involvement of key molecular components from these three pathways while defying full classification under any single one. Its introduction marks a significant breakthrough in understanding cell death mechanisms, establishing a new framework to study the role of cell death in diseases. Recent studies have demonstrated the crucial role of PANoptosis in the progression of various diseases, including infections, inflammation, and cancer. This emerging concept offers novel perspectives for exploring the impact of cell death on disease mechanisms and potential therapeutic interventions.

Figure 1 Molecular Mechanisms of Pyroptosis, Apoptosis, Necroptosis, and PANoptosis. (1) Pyroptosis: Pyroptosis involves inflammasome formation, typically the Nucleotide-Binding Domain, Leucine-Rich Repeat, and Pyrin Domain Containing Protein (NLRP) 3 inflammasome comprising the NLRP3 sensor, Apoptosis-Associated Speck-Like Protein Containing a CARD (ASC) adaptor, and Pro-Caspase-1. Activated inflammasomes cleave Caspase-1, which then processes Interleukin (IL)-1β/18 and Gasdermin D (GSDMD). The N-terminal fragment of GSDMD oligomerizes on the plasma membrane, forming pores that execute cell death. (2) Apoptosis: Apoptosis operates via extrinsic and intrinsic pathways: In the extrinsic pathway, external stimuli activate death receptors, forming the Fas-Associated protein with Death Domain (FADD)/Caspase-8/Receptor-Interacting Protein Kinase (RIPK1) complex. This activates Caspase-8, which cleaves Caspase-3/-7 to execute cell death. In the intrinsic pathway, mitochondrial outer membrane permeability changes, releasing cytochrome C. This promotes apoptosome formation, activating Caspase-9, which subsequently cleaves Caspase-3/-7 to execute cell death. (3) Necroptosis: When Caspase-8 is absent or inhibited, necroptosis occurs. Tumor Necrosis Factor-alpha (TNF-α) binding to its receptor forms the necrosome, containing RIPK1 and RIPK3. Phosphorylated RIPK1/RIPK3 recruits and phosphorylates Mixed Lineage Kinase Domain-Like Protein (MLKL), which disrupts membrane integrity to induce cell death. (4) PANoptosis: PANoptosis is mediated by the PANoptosome, integrating components from other cell death pathways. Pathogens activate sensors such as Z-DNA binding protein 1 (ZBP1), AIM2, RIPK1, or NLRP12, forming a PANoptosome complex that includes inflammasome components (eg, ASC), complex II elements (eg, Caspase-8), and necrosome components (eg, RIPK3). IRF1 further facilitates this process. The PANoptosome induces cell death by activating CASP1 (cleaving GSDMD and IL-1β/18), Caspase-3/-7 (cleaving Gasdermin E), and phosphorylating MLKL.

PANoptosis plays a critical role in maintaining bodily homeostasis by eliminating pathogen-infected cells and triggering immune responses through the release of inflammatory cytokines, thereby protecting the host from infections. For instance, during viral infections, Z-DNA binding protein 1 (ZBP1) detects viral RNA and activates the PANoptosome, leading to the death of infected cells and limiting viral replication and spread.50 PANoptosis also contributes to the removal of damaged, aging, or cancerous cells, essential for tissue health and tumor suppression.51 In the context of disease, PANoptosis plays a dual role. It is involved in various pathological processes, including infectious diseases, inflammatory disorders, cancer, cardiovascular conditions, and neurodegenerative diseases. For example, in infectious diseases, pathogens such as bacteria, viruses, and fungi can induce PANoptosis, limiting pathogen spread. However, excessive PANoptosis can exacerbate tissue damage and inflammation, worsening disease symptoms.49,52 The process of PANoptosis also plays a crucial role in promoting intervertebral disc degeneration.53 In inflammatory diseases, PANoptosis-mediated immune cell death releases inflammatory cytokines, intensifying the inflammatory response.54 Simultaneously, it regulates immune cell activity and function, influencing immune modulation during inflammation.55,56 In cancer, PANoptosis also exhibits dual effects. Targeting PANoptosis-related molecules can induce cancer cell death and inhibit tumor growth. However, cancer cells may exploit PANoptosis activation to evade immune surveillance and resist chemotherapy.57

Molecular Mechanisms of PANoptosis

PANoptosome

The PANoptosome is the central molecular platform for executing PANoptosis, enabling cells to initiate PANoptosis instead of a single “classical” RCD pathway. The assembly of the PANoptosome involves three main categories of proteins: (1) sensors for Pathogen-Associated Molecular Patterns (PAMPs) or Damage-Associated Molecular Patterns (DAMPs), such as ZBP1, Absent in Melanoma 2 (AIM2), and Nucleotide-Binding Domain, Leucine-Rich Repeat, and Pyrin Domain Containing Protein (NLRP) 3; (2) adaptor proteins, including Apoptosis-Associated Speck-Like Protein Containing a CARD (ASC) and Fas-Associated protein with Death Domain (FADD); and (3) catalytic effectors, such as RIPK1, RIPK3, Caspase-1, and Caspase-8.52,58

Several PANoptosomes have been identified, including ZBP1-PANoptosome, AIM2-PANoptosome, RIPK1-PANoptosome, and NLRP12-PANoptosome.59–62 ZBP1 was the first PANoptosis sensor identified. It detects Z-DNA or Z-RNA during viral infections, triggering cell death signals. ZBP1 uses its Zα domain to bind the Receptor-Interacting Protein Homotypic Interaction Motif (RHIM) domain of RIPK3, facilitating PANoptosome assembly. AIM2 initiates PANoptosis by detecting cytosolic DNA fragments through its Hematopoietic Interferon-inducible Nuclear proteins with a 200-amino acid repeat (HIN200) domain. It recruits ASC, which activates Caspase-1, driving downstream cell death processes. RIPK1 acts as a central hub for PANoptosome formation and integrates cell death signals. It activates Caspase-8 via FADD to induce apoptosis or interacts with RIPK3 to drive necroptosis or pyroptosis, depending on the signaling context and molecular interactions. NLRP12 senses heme or PAMPs via its Leucine-Rich Repeat (LRR) domain and activates the Caspase-8/RIPK3 axis, assembling the PANoptosome and triggering inflammatory and cell death responses.63 The PANoptosome represents a highly flexible and efficient cell death platform, orchestrated through the precise cooperation of key proteins. Its dynamic adaptability allows cells to respond to diverse stressors with tailored cell death mechanisms.

Signaling Pathways of PANoptosis

The initiation of PANoptosis relies on a complex network of upstream signals. These networks detect PAMPs and DAMPs to activate multiple signaling pathways that induce cell death. Pattern recognition receptors (PRRs), such as Toll-like receptors (TLRs), RIG-I-like receptors (RLRs), nucleotide-binding oligomerization domain-like receptors (NLRs), and ZBP1, are key sensors for PANoptosis.64,65 PRRs recognize viral RNA and other PAMPs, such as TLR7 detecting Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV) and RIG-I recognizing influenza virus (IAV) or hepatitis C virus (HCV) RNA.6570 The amplification of these signals often depends on type I interferon signaling pathways, which induce ZBP1 expression through the Janus Kinase (JAK)/Signal Transducer and Activator of Transcription (STAT) pathway. ZBP1, in turn, recognizes viral RNA via its Zα domain, activating the Nucleotide-Binding Domain, Leucine-Rich Repeat, and NLRP3 inflammasome and triggering PANoptosis.50 Cytokine signals, such as tumor necrosis factor-alpha (TNF-α) and interferon-gamma (IFN-γ), can also induce nitric oxide production via the JAK/STAT1/Interferon Regulatory Factor(IRF) 1 axis, activating the caspase-8/FADD complex and driving PANoptosis.54 Additionally, TLRs directly activate NLRP3 and AIM2 inflammasomes and regulate other cell death signals, such as the Stimulator of Interferon Genes (STING) pathway, which promotes ZBP1 and AIM2 activation by inducing IRF3 expression.71 The Transforming Growth Factor-β-Activated Kinase 1 (TAK1) signaling pathway plays a dual role in PANoptosis: it maintains NLRP3 in a resting state but triggers RIPK1-dependent or independent PANoptosis when inhibited or absent.48,72,73 Oxidative stress signals also link mitochondrial dysfunction to PANoptosis by activating the NLRP3 inflammasome.74–76 Through these interconnected networks, molecules and signaling pathways collectively integrate upstream signals to coordinate the induction of PANoptosis.

At the downstream level, PANoptosis integrates and executes multiple RCD pathways through the coordinated activation of various signaling cascades. The caspase signaling pathway plays a pivotal role, with Caspase-1, Caspase-8, and effector caspases (Caspase-3/7) being key mediators. These caspases not only activate inflammasomes but also directly execute apoptosis and necroptosis.77 The RIPK1/RIPK3/MLKL pathway, central to necroptosis, involves the formation of the necrosome complex through RIPK1-RIPK3 interactions. This activates MLKL, which oligomerizes upon phosphorylation, forming membrane pores that disrupt membrane integrity and lead to cell death.78 Pyroptosis is mediated by inflammasomes such as NLRP3 and AIM2, which detect viral invasion and activate Caspase-1. Caspase-1 cleaves GSDMD, whose pore formation causes cell lysis while releasing pro-inflammatory cytokines like IL-1β and IL-18.79,80 A defining feature of PANoptosis is the crosstalk between these pathways. For instance, the ZBP1-PANoptosome complex detects viral RNA and recruits the NLRP3 inflammasome to activate Caspase-1. Simultaneously, it triggers the RIPK1/RIPK3 axis to activate Caspase-8, integrating pyroptosis, necroptosis, and apoptosis into a unified response.81 Additionally, PANoptosis relies on key effector molecules such as GSDMs and MLKL, which execute membrane disruption in pyroptosis and necroptosis, respectively, ultimately resulting in cell death. These interwoven signaling pathways form a tightly coordinated network, regulated through platforms like the PANoptosome, ensuring efficient execution of PANoptosis.

PANoptosis in Sepsis

PANoptosis Related Genes and Sepsis

Research on PANoptosis-related genes (PRGs) in sepsis has identified 16 genes that determine sepsis subtypes. By examining molecular clustering and prognostic features based on PANoptosis, the immune landscape and prognosis of sepsis patients can be predicted. It was also found that PRGs exhibit different expression patterns in various immune cells.82 One study confirmed that PANoptosis-related genes show significant predictive power for sepsis, with the differential expression of these genes being strongly associated with sepsis prognosis and leading to organ dysfunction.83 Another study found that PRGs are highly useful in the early detection of pediatric septic shock. This is crucial for understanding the role of immune cell infiltration in the pathophysiology of pediatric septic shock, though further confirmation through basic and clinical research is needed.84 A diagnostic prediction model based on PRGs was able to effectively distinguish between septic ARDS patients and general sepsis patients. NDRG1 was identified as a potential therapeutic target, and subsequent research in a sepsis mouse model confirmed that inhibiting NDRG1 could alleviate lung injury.85 A study combining bioinformatics analysis and experimental validation suggested that four PANoptosis genes—CD14, GSDMD, IL-1β, and Fas—are highly involved in the immune response and multiple inflammatory pathways during sepsis-induced acute lung injury.86 In addition to studies on the association between PANoptosis-related genes and sepsis, PANoptosis is also linked to sepsis through its role in exacerbating inflammatory responses, impairing immune function, and contributing to organ dysfunction (Table 1).

Table 1 Studies Related to PANoptosis in Sepsis

Effects of PANoptosis on Inflammatory Responses

PANoptosis triggers the release of inflammatory cytokines through multiple mechanisms, leading to inflammatory storms. Under conditions of infection or cellular stress, intracellular PRRs, such as ZBP1 and NOD-like receptor family CARD domain-containing protein 4 (NLRC4), detect PAMPs or DAMPs. This activates inflammasomes, including NLRP3, which leads to the autocleavage and activation of Caspase-1. Activated Caspase-1 catalyzes the maturation and secretion of interleukin (IL)-1β and IL-18 while inducing GSDMD cleavage to form membrane pores that drive pyroptotic cell death.81,102 These inflammatory cytokines amplify the local and systemic inflammatory responses, playing a significant role in conditions such as infections, trauma, and autoimmune diseases.103 Second, the combined action of TNF-α and IFN-γ has been shown to induce PANoptosis by activating the IRF1 and nitric oxide synthase 2 (NOS2) axis. This triggers the cleavage of GSDM family members, including GSDMD and GSDME, intensifying cell death and cytokine release.54 Additionally, the necroptosis pathway mediated by RIPK1/RIPK3 results in the phosphorylation and activation of MLKL. Activated MLKL disrupts membrane integrity by forming pores, and releasing cellular contents, including inflammatory cytokines.104 Caspase-8, apart from its role in apoptosis, also contributes to pyroptosis and necroptosis, influencing the production and release of inflammatory cytokines through its interactions with other apoptosis-related proteins.21,105 The excessive production of these inflammatory cytokines not only exacerbates local inflammatory responses but can also spread systemically, resulting in a full-scale inflammatory storm.

PANoptosis-induced activation of inflammasome and necroptosis leads to the release of inflammatory cytokines such as TNF-α, IL-1β, and IL-6, which are elevated in the serum of sepsis patients.106 These cytokines amplify inflammation, exacerbate tissue damage, and create a vicious cycle. Excessive production of inflammatory cytokines such as IL-1β, IL-6, and TNF-α during a cytokine storm not only directly damages tissues but also disrupts vascular barrier function. These cytokines activate endothelial cells, increase vascular permeability, and promote thrombosis, leading to severe complications like pulmonary edema and ARDS.107 Additionally, they activate immune cells, particularly neutrophils, which release large amounts of reactive oxygen species (ROS) and neutrophil extracellular traps (NETs), further exacerbating tissue damage.108,109 Research has demonstrated the critical role of ZBP1, a key component of the PANoptosis in sepsis.110 The activation of PANoptosis leads to the release of inflammatory cytokines, contributing to AKI and exacerbating renal dysfunction.98,111 PANoptosis also drives acute lung injury (ALI) through pro-inflammatory pathways and inhibition of PANoptosis has been shown to alleviate ALI symptoms.94–96,99,100 Moreover, PANoptosis has been implicated in sepsis-associated intestinal dysfunction, myocardial suppression, liver dysfunction, and the development of multiple organ dysfunction syndrome (MODS), significantly increasing mortality in sepsis patients.54,97,101,111 In addition to pro-inflammatory cytokines, PANoptosis alters the secretion of anti-inflammatory cytokines. IL-10 and Transforming Growth Factor-beta (TGF-β) are known for their immunosuppressive and anti-inflammatory roles. Increased TGF-β expression during PANoptosis in gastric cancer, indicates the complexity of this process.112 Understanding the mechanisms of PANoptosis and its role in cytokine storms is crucial for developing effective therapeutic strategies to mitigate the severe outcomes of sepsis.

Effects of PANoptosis on Immune Cells

PANoptosis occurs in various pathological conditions, including sepsis and autoimmune diseases, and is often associated with a reduction in the number of immune cells. For instance, in sepsis, the numbers and functions of immune cells such as T lymphocytes, B lymphocytes, monocytes, and macrophages are significantly diminished due to PANoptosis.54 These cells are critical for immune responses and inflammation regulation, and their depletion impairs immune function, negatively affecting patient outcomes.25,106 PANoptosis not only reduces the overall number of immune cells but also impacts their differentiation. In systemic lupus erythematosus (SLE), for example, regulatory T cells (Tregs) decrease, while effector T cells increase, leading to immune imbalance. CD8+ T cells, known for their cytotoxic activity, often exhibit dysfunction in SLE patients, increasing their susceptibility to infections.113 Studies suggest that PANoptosis in T cells may contribute to the immune dysregulation seen in SLE.114 Antigen-presenting cells (APCs), such as macrophages and dendritic cells (DCs), are also affected by PANoptosis. DC depletion due to PANoptosis impairs antigen uptake, processing, and presentation, hindering T cell activation and immune responses.115 For example, DCs induce cytokine production and PANoptosis while presenting cancer antigens to T cells.116 In sepsis, DC numbers in peripheral blood are reduced, compromising their antigen-presenting function and impairing T cell proliferation and differentiation.117,118 B cell subsets are also selectively depleted in sepsis, accompanied by selective Caspase-1, 3, 8 expression.119 The Caspase family is known to be closely associated with PANoptosis.105,120

The mechanisms underlying immune cell exhaustion or dysfunction are complex and multifaceted. Dysregulation of cell death signaling pathways plays a crucial role. Under stimuli such as infection or tissue damage, excessive activation of inflammasomes increases the sensitivity of immune cells to PANoptosis, accelerating immune cell depletion.54 Key molecules in the inflammasome signaling pathway, including NLRP3, ASC, and Caspase-1, are often implicated. Mutations or abnormal expression of these molecules can disrupt inflammasome pathways, impairing immune cell survival and function. For instance, NLRP3 mutations are strongly associated with autoimmune diseases and can lead to hyperactivation of inflammasomes, resulting in immune cell exhaustion.52,121 Immune cell metabolic dysregulation is another contributing factor. The metabolic state of immune cells is essential for their function and survival. During PANoptosis, immune cell energy metabolism may become disrupted, leading to reduced Adenosine Triphosphate (ATP) production and mitochondrial dysfunction.74,75,122 As the primary energy source for cellular activities, ATP depletion affects immune cell proliferation, differentiation, and functionality. Mitochondrial dysfunction further exacerbates the situation by increasing ROS production, which damages cellular structures and functions. PANoptosis may also impact the synthesis and breakdown of metabolites such as amino acids and nucleotides, which are critical for protein synthesis, DNA replication, and repair.123,124 Dysregulation of these processes can impair immune cell functions and promote exhaustion. In addition, changes in the immune microenvironment play a significant role. PANoptosis-induced cytokine storms result in the excessive release of inflammatory cytokines, altering the immune cell microenvironment.54 This suppresses immune cell proliferation and differentiation while accelerating exhaustion.125 PANoptosis may also lead to immune cell exhaustion, such as in T cells, B cells, and macrophages, thereby affecting their interactions. Immune cell interactions are crucial for maintaining immune homeostasis, and their disruption can lead to functional impairment and exhaustion.

PANoptosis in Organ Dysfunction

In addition to contributing to organ damage through cytokine storms, PANoptosis leads to multi-organ dysfunction via other complex mechanisms (Figure 2). In cardiomyocytes, PANoptosis is activated by PRRs such as ZBP1, which detect viral-derived Z-nucleic acids or endogenous nucleic acids. This triggers inflammatory cell death, reducing cardiomyocyte numbers and releasing pro-inflammatory mediators that exacerbate local inflammation and impair cardiac function.101 In sepsis-induced ALI, PANoptosis is driven by the activation of ZBP1, RIPK1, NLRP3 inflammasomes, and STING pathways. This process induces metabolic dysfunction and oxidative stress, exacerbates inflammation and tissue damage, disrupts lung barrier integrity, increases vascular permeability, and leads to pulmonary edema, severely impairing gas exchange. Inhibition of PANoptosis with drugs can reduce inflammation and alleviate ALI.94–96,99,100 PANoptosis also plays a critical role in AKI. In renal tubular epithelial cells, PANoptosis is primarily mediated by the AIM2-PANoptosome. The formation of the PANoptosome consisting of ZBP1, ASC, and RIPK1 was involved in the development of kidney injury.98,111 Loss of tubular epithelial cells compromises tubular function, disrupting electrolyte balance and waste excretion, thereby exacerbating kidney failure. Studies have shown that PANoptosis plays a role in sepsis-associated encephalopathy. TLR9 activates PANoptosis through the p38 mitogen-activated protein kinase signaling pathway, leading to the progression of SAE and increased mortality.97 In summary, PANoptosis significantly impacts organ function by inducing various forms of cell death in sepsis, ultimately worsening patient outcomes.

Figure 2 The immunogenic effects of PANoptosis and its role in sepsis. In sepsis, PANoptosis plays a critical role. It triggers an inflammatory cytokine storm, leading to the massive release of inflammatory factors such as IL-1β, IL-6, and TNF-α into the peripheral blood. It also causes a significant reduction in the number of immune cells in the peripheral blood and spleen, accompanied by functional impairment. Additionally, PANoptosis contributes to multi-organ dysfunction, with conditions such as myocardial depression, lung injury, and kidney injury being closely associated with its effects.

Therapeutic Strategies Targeting PANoptosis

Drug Development and Applications

Efforts to develop chemical drugs targeting key molecules or signaling pathways of PANoptosis are ongoing. Significant progress has been made in designing inhibitors for multiple pathways, reducing cell death.126

Several small-molecule compounds targeting RIPK1 and RIPK3 have been developed to inhibit necroptosis. Necrostatin and its derivatives (Necrostatin-3, Dimethyl Oxazolone, Necrostatin-4 Series) are specific RIPK1 inhibitors with significant effects in necroptotic diseases and inflammatory conditions.127–131 Other RIPK1 inhibitors include Sunitinib, Tozasertib, Butylated Hydroxyanisole, KW-244, and Benzoxazepine derivative GSK2982772.127,132–135 Compounds such as GNF-7, GSK481, and ZJU-37 target both RIPK1 and RIPK3 to suppress necroptosis.136–138 Molecules like GSK840, GSK843, GSK872, and AZD5423 regulate the RIPK3/MLKL pathway, further expanding the drug repertoire for necroptosis modulation.139,140 Disulfiram, Dimethyl fumarate (DMF), necrosulfonamide (NSA), and Bioorthogonally ACtivatable Base editor target GSDMD and GSDME to suppress pyroptosis in diseases including sepsis.94,141–144 VX765 and Acetyl-Tyrosine-Valine-Alanine-Aspartic Acid-Chloromethylketone (Ac-YVAD-CMK) inhibit Caspase-1 activity.145,146 Additionally, Dickkopf-related protein 1 blocks GSDMD, Caspase-3, and RIPK3 to suppress PANoptosis.147

Drugs targeting PANoptosome components have shown promise in reducing cell death and inflammation. Melatonin suppresses PANoptosis by reducing the expression of key components such as NLRP3, ASC, caspase-1, GSDMD, RIPK1, RIPK3, and MLKL.148 Cucurbitacin E modulates PANoptosis in Adenoid Cystic Carcinoma cells by interacting with the PANoptosome in a ZBP1-dependent manner.149 Penehyclidine hydrochloride (PHC) decreases myocardial enzyme release and inhibits ZBP1 to alleviate PANoptosis.150 MiR-29a-3p down-regulates ZBP1, GSDMD caspase-3, caspase-8, and MLKL Reduces alveolar epithelial cell PANoptosis and down-regulates inflammatory factor expression such as TNF-α, IL-1β and IL-6 in the lung.95 OLT1177, MCC950, Tranilast, β-hydroxybutyrate, and 3,4-Methylenedioxy-β-Nitrostyrene (MNS) effectively inhibit NLRP3 inflammasome activation.151–156 J114 disrupts NLRP3/AIM2 interactions with ASC, inhibiting inflammasome oligomerization.157 Obovatol suppresses NLRP3, AIM2 inflammasome activation and mitochondrial ROS production.158 AR1D1 interacts with ZBP1’s Zα2 domain to block its interaction with RIPK3 to inhibit PANoptosis, and drugs targeting this target are worth developing.159 Drugs targeting inflammatory signaling pathways also inhibit PANoptosis. TNF-α and IFN-γ neutralizing antibodies protect against sepsis and cytokine storm-related damage by preventing their synergistic effects.125 Emricasan, a broad-spectrum caspase inhibitor, reduces cell death and protects tissues in multiple diseases.160,161 Inhibitors of the JAK/STAT signaling pathway are considered potential therapeutic targets for PANoptosis. Among them, the JAK1/2 inhibitor Baricitinib, approved for Coronavirus Disease 2019 treatment, has demonstrated significant improvements in patient survival rates.162,163 Myricetin alleviates disseminated intravascular coagulation (DIC) in sepsis by reducing platelet activation and PANoptosis.93 1-Methoxy-5-Methylphenazinium Methyl Sulfate (MPMS) and DMF inhibit mitochondrial DNA oxidation and PANoptosome formation, mitigating sepsis-related pathology.111

While these drugs have shown efficacy in preclinical models, challenges remain in translating them into clinical use. Diseases may require tailored combination therapies to target specific pathways effectively. Long-term effects and potential interference with normal immune responses need careful evaluation. For example, while TNF-α and IFN-γ antibodies reduce inflammation, their immunosuppressive effects may increase infection risk. The NLRP3 inhibitor MCC950 has demonstrated strong anti-inflammatory effects across various inflammatory disease models, effectively reducing cell death.152 Inhibitors targeting key PANoptosome components, such as Necrostatin, have shown therapeutic potential in models of ischemia-reperfusion injury, neurodegenerative disorders, and lung injury. Promising results in animal models may not directly apply to humans, necessitating rigorous clinical trials. In conclusion, although substantial progress has been made in PANoptosis-targeted drug development, further research is needed to optimize therapies for safe and effective clinical application.

The Potential Role of Traditional Chinese Medicine

In recent years, traditional Chinese medicine (TCM) and its active components have shown significant potential in regulating PANoptosis due to their multi-target and multi-pathway mechanisms. For instance, XiaoChaiHu Decoction (XCHD) has demonstrated protective effects in sepsis-induced cardiomyopathy. Active components of XCHD have been identified through network pharmacology as potential targets for PANoptosis, significantly suppressing LPS-induced myocardial inflammatory cytokines (eg, IL-6, IL-1β, TNF-α) and reducing PANoptosis-related gene expression.101 Similarly, ursodeoxycholic acid (UDCA) alleviates sepsis-induced ALI by blocking the STING pathway, accompanied by suppression of PANoptosis.100 Other TCM components, such as baicalin and ginsenosides, exhibit antioxidant, anti-inflammatory, and anti-cell exhaustion effects. These properties help mitigate sepsis-induced tissue damage and improve prognosis. Baicalin blocks mitochondrial DNA release and oxidation, reducing intracellular ROS and mitochondrial ROS during PANoptosis. It also inhibits ZBP1-related PANoptosome assembly. In macrophages (Kupffer cells), PANoptotic signaling activation leads to liver damage in hemophagocytic lymphohistiocytosis (HLH) mouse models, which baicalin effectively mitigates by suppressing PANoptosis.59 Ginsenosides suppress NLRP3 inflammasome activation, reduce ASC oligomerization, and downregulate apoptosis, inflammation, and oxidative stress in sepsis models.164,165 Duhuo Jisheng Decoction may exert its effects in intervertebral disc degeneration by inhibiting the processes of apoptosis, necrosis, and pyroptosis.166 Scutellarin, another TCM component, protects against systemic inflammation, multi-organ damage, and PANoptosis in HLH mice by inhibiting mitochondrial damage and mtROS generation.122 Similarly, Shengxian Decoction (SXD) alleviates bleomycin-induced pulmonary fibrosis by inhibiting ZBP1-mediated PANoptosis. It targets pyroptosis, apoptosis, and necroptosis, effectively delaying or reversing early pathological changes.167 Achyranthes aspera extract, which downregulates genes and pathways related to DNA damage, oxidative stress, inflammation, and PANoptosis, while promoting survival-related signaling.168 Dachengqi Decoction (DDG), which inhibits excessive inflammation and PANoptosis, particularly upstream regulators like ZBP1 and RIPK1, to protect against LPS-induced ALI.96 Cranberry-derived exosomes significantly suppress PANoptosis in mice by suppressing ROS release and cytokine expression.169

In summary, TCM offers a promising approach for regulating PANoptosis and mitigating inflammation-related diseases by targeting multiple pathways and key molecules.

Conclusion and Future Perspectives

Sepsis, a critical global public health challenge, is closely linked to RCD. As an emerging form of RCD, PANoptosis plays a pivotal role in the pathophysiology of sepsis, influencing inflammation, immune cell function, and multi-organ damage. Targeting PANoptosis pathways has thus become a promising direction for developing new therapeutic strategies for sepsis. Both chemical drugs targeting key molecules or signaling pathways of PANoptosis and traditional Chinese medicines with multi-target, multi-pathway mechanisms have shown potential in regulating PANoptosis.

Despite these advances, the precise regulatory mechanisms of PANoptosis remain incompletely understood. Critical questions, such as the specific triggers of PANoptosis under different conditions, the assembly process of the PANoptosome, and its interactions with other cell death pathways, require further investigation. Further research is also needed to identify more precise molecular targets. The development of effective PANoptosis-targeted drugs is another priority. Although some compounds have shown potential in inhibiting PANoptosis, their mechanisms of action remain unclear, and their safety and efficacy in clinical settings need validation. Future research should focus on identifying high-efficacy, highly specific PANoptosis inhibitors and evaluating their therapeutic potential through animal models and clinical trials. Additionally, integrating advanced technologies such as single-cell transcriptomics and bioinformatics will provide deeper insights into the role of PANoptosis in personalized medicine. This approach could pave the way for precision-targeted therapies, offering tailored treatment options for sepsis patients. In conclusion, as our understanding of PANoptosis continues to grow and innovative technologies advance, targeting PANoptosis holds the potential to significantly improve sepsis treatment outcomes and patient prognosis, offering hope in addressing this global health challenge.

Abbreviations

Ac-YVAD-CMK, Acetyl-Tyrosine-Valine-Alanine-Aspartic Acid-Chloromethylketone; ALI, Acute Lung Injury; AIM2, Absent in Melanoma 2; APCs, Antigen-Presenting Cells; ARDS, Acute Respiratory Distress Syndrome; ASC, Apoptosis-Associated Speck-Like Protein Containing a CARD; ATP, Adenosine Triphosphate; Bcl-2, B-cell lymphoma 2; Bax, Bcl-2-associated X protein; DCs, Dendritic Cells; DAMPs, Damage-Associated Molecular Patterns; DDG, Dachengqi Decoction; DMF, Dimethyl fumarate; DIC, Disseminated Intravascular Coagulation; FADD, Fas-Associated protein with Death Domain; GSDMD, Gasdermin D; GSDME, Gasdermin E; HIV, Human Immunodeficiency Virus; HLH, Hemophagocytic Lymphohistiocytosis; HIN200, Hematopoietic Interferon-inducible Nuclear proteins with a 200-amino acid repeat; IFN-γ, Interferon-gamma; IL, Interleukin; IRF, Interferon Regulatory Factor; IAV, Influenza Virus; JAK, Janus Kinase; LRR, Leucine-Rich Repeat; MLKL, Mixed Lineage Kinase Domain-Like Protein; MODS, Multiple Organ Dysfunction Syndrome; MPMS, 1-Methoxy-5-Methylphenazinium Methyl Sulfate; MNS, 3,4-Methylenedioxy-β-Nitrostyrene; NLRC4, NOD-like receptor family CARD domain-containing protein 4; NLRP, Nucleotide-Binding Domain, Leucine-Rich Repeat, and Pyrin Domain Containing Protein; NOS2, Nitric Oxide Synthase 2; NSA, Necrosulfonamide; NLRs, Nucleotide-Binding Oligomerization Domain-like Receptors; PAMPs, Pathogen-Associated Molecular Patterns; RCD, Regulated cell death; PHC, Penehyclidine hydrochloride; PRRs, Pattern Recognition Receptors; RHIM, Receptor-Interacting Protein Homotypic Interaction Motif; RIG-I, Retinoic Acid-Inducible Gene I; RIPK1, Receptor-Interacting Protein Kinase 1; RIPK3, Receptor-Interacting Protein Kinase 3; RLR, RIG-I-like receptors; ROS, Reactive Oxygen Species; SAE, sepsis-associated encephalopathy; SLE, Systemic Lupus Erythematosus; STAT, Signal Transducer and Activator of Transcription; STING, Stimulator of Interferon Genes; SXD, Shengxian Decoction; TAK1, Transforming Growth Factor-β-Activated Kinase 1; TCM, Traditional Chinese Medicine; TGF-β, Transforming Growth Factor-beta; TNF-α, Tumor Necrosis Factor-alpha; TLRs, Toll-like receptors; Tregs, Regulatory T Cells; UDCA, Ursodeoxycholic Acid; XCHD, XiaoChaiHu Decoction; ZBP1, Z-DNA binding protein 1.

Acknowledgments

We appreciate all authors whose publications could be included in our review.

Author Contributions

All authors have read and agreed to the published version of the manuscript. Qi SY. and Wu QQ. searched and collected the literatures. Qi SY. wrote the manuscript and Wu QQ. finished the drawing, revision, and response to reviewers’ comments of the paper. Kang ZF., Bai XJ., and Li ZF., evaluated the theoretical basis and conclusions. Dong XJ., and Cheng Jing. supervised and reviewed the manuscript. All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work. Qiqi Wu and Siyuan Qi share senior authorship.

Funding

National Natural Science Foundation of China (No. 82202387); Natural Science Foundation of Department of Science and Technology of Hubei Province (No. 2022CFB278).

Disclosure

The authors declare no conflict of interests.

References

1. Singer M, Deutschman CS, Seymour CW, et al. The third international consensus definitions for sepsis and septic shock (sepsis-3). JAMA. 2016;315(8):801–810. doi:10.1001/jama.2016.0287

2. Zhang YY, Ning BT. Signaling pathways and intervention therapies in sepsis. Signal Transduct Target Ther. 2021;6(1):407. doi:10.1038/s41392-021-00816-9

3. Cecconi M, Evans L, Levy M, Rhodes A. Sepsis and septic shock. Lancet. 2018;392(10141):75–87. doi:10.1016/S0140-6736(18)30696-2

4. Hollenberg SM, Singer M. Pathophysiology of sepsis-induced cardiomyopathy. Nat Rev Cardiol. 2021;18(6):424–434. doi:10.1038/s41569-020-00492-2

5. Song J, Fang X, Zhou K, Bao H, Li L. Sepsis‑induced cardiac dysfunction and pathogenetic mechanisms (Review). Mol Med Rep. 2023;28(6). doi:10.3892/mmr.2023.13114

6. Li Y, Yu J, Li R, Zhou H, Chang X. New insights into the role of mitochondrial metabolic dysregulation and immune infiltration in septic cardiomyopathy by integrated bioinformatics analysis and experimental validation. Cell Mol Biol Lett. 2024;29(1):21. doi:10.1186/s11658-024-00536-2

7. Sheng SY, Li JM, Hu XY, Wang Y. Regulated cell death pathways in cardiomyopathy. Acta Pharmacol Sin. 2023;44(8):1521–1535. doi:10.1038/s41401-023-01068-9

8. Meng S, Liu J, Wang Z, et al. Inhibition of Golgi stress alleviates sepsis-induced cardiomyopathy by reducing inflammation and apoptosis. Int Immunopharmacol. 2024;133:112103. doi:10.1016/j.intimp.2024.112103

9. Chaudhuri D, Nei AM, Rochwerg B, et al. Focused update: guidelines on use of corticosteroids in sepsis, acute respiratory distress syndrome, and community-acquired pneumonia. Crit Care Med. 2024;52(5):e219–e233. doi:10.1097/CCM.0000000000006172

10. Chang Y, Yoo HJ, Kim SJ, et al. A targeted metabolomics approach for sepsis-induced ARDS and its subphenotypes. Crit Care. 2023;27(1):263. doi:10.1186/s13054-023-04552-0

11. Poston JT, Koyner JL. Sepsis associated acute kidney injury. BMJ. 2019;364:k4891.

12. Manrique-Caballero CL, Del Rio-Pertuz G, Gomez H. Sepsis-associated acute kidney injury. Crit Care Clin. 2021;37(2):279–301. doi:10.1016/j.ccc.2020.11.010

13. van der Slikke EC, Star BS, van Meurs M, Henning RH, Moser J, Bouma HR. Sepsis is associated with mitochondrial DNA damage and a reduced mitochondrial mass in the kidney of patients with sepsis-AKI. Crit Care. 2021;25(1):36. doi:10.1186/s13054-020-03424-1

14. Zou R, Tao J, Qiu J, et al. DNA-PKcs promotes sepsis-induced multiple organ failure by triggering mitochondrial dysfunction. J Adv Res. 2022;41:39–48. doi:10.1016/j.jare.2022.01.014

15. Piva S, Bertoni M, Gitti N, Rasulo FA, Latronico N. Neurological complications of sepsis. Curr Opin Crit Care. 2023;29(2):75–84. doi:10.1097/MCC.0000000000001022

16. Bao W, Xing H, Cao S, et al. Neutrophils restrain sepsis associated coagulopathy via extracellular vesicles carrying superoxide dismutase 2 in a murine model of lipopolysaccharide induced sepsis. Nat Commun. 2022;13(1):4583. doi:10.1038/s41467-022-32325-w

17. Liang H, Song H, Zhang X, et al. Metformin attenuated sepsis-related liver injury by modulating gut microbiota. Emerg Microbes Infect. 2022;11(1):815–828. doi:10.1080/22221751.2022.2045876

18. Galluzzi L, Bravo-San Pedro JM, Vitale I, et al. Essential versus accessory aspects of cell death: recommendations of the NCCD 2015. Cell Death Differ. 2015;22(1):58–73. doi:10.1038/cdd.2014.137

19. Fleisher TA. Apoptosis. Ann Allergy Asthma Immunol. 1997;78(3):245–249;quiz249–250. doi:10.1016/S1081-1206(10)63176-6

20. Kovacs SB, Miao EA. Gasdermins: effectors of pyroptosis. Trends Cell Biol. 2017;27(9):673–684. doi:10.1016/j.tcb.2017.05.005

21. Bertheloot D, Latz E, Franklin BS. Necroptosis, pyroptosis and apoptosis: an intricate game of cell death. Cell Mol Immunol. 2021;18(5):1106–1121.

22. Shi C, Cao P, Wang Y, et al. PANoptosis: a cell death characterized by pyroptosis, apoptosis, and necroptosis. J Inflamm Res. 2023;16:1523–1532. doi:10.2147/JIR.S403819

23. Qi X, Yu Y, Sun R, et al. Identification and characterization of neutrophil heterogeneity in sepsis. Crit Care. 2021;25(1):50. doi:10.1186/s13054-021-03481-0

24. Cao C, Yu M, Chai Y. Pathological alteration and therapeutic implications of sepsis-induced immune cell apoptosis. Cell Death Dis. 2019;10(10):782. doi:10.1038/s41419-019-2015-1

25. Venet F, Monneret G. Advances in the understanding and treatment of sepsis-induced immunosuppression. Nat Rev Nephrol. 2018;14(2):121–137. doi:10.1038/nrneph.2017.165

26. Girardot T, Rimmelé T, Venet F, Monneret G. Apoptosis-induced lymphopenia in sepsis and other severe injuries. Apoptosis. 2017;22(2):295–305. doi:10.1007/s10495-016-1325-3

27. Galluzzi L, Vitale I, Aaronson SA, et al. Molecular mechanisms of cell death: recommendations of the nomenclature committee on cell death 2018. Cell Death Differ. 2018;25(3):486–541. doi:10.1038/s41418-017-0012-4

28. Zhu CL, Wang Y, Liu Q, et al. Dysregulation of neutrophil death in sepsis. Front Immunol. 2022;13:963955. doi:10.3389/fimmu.2022.963955

29. Briassoulis G, Briassoulis P, Ilia S, Miliaraki M, Briassouli E. The anti-oxidative, anti-inflammatory, anti-apoptotic, and anti-necroptotic role of zinc in COVID-19 and sepsis. Antioxidants. 2023;12(11). doi:10.3390/antiox12111942

30. Zheng X, Chen W, Gong F, Chen Y, Chen E. The role and mechanism of pyroptosis and potential therapeutic targets in sepsis: a review. Front Immunol. 2021;12:711939. doi:10.3389/fimmu.2021.711939

31. Jiao Y, Zhang T, Zhang C, et al. Exosomal miR-30d-5p of neutrophils induces M1 macrophage polarization and primes macrophage pyroptosis in sepsis-related acute lung injury. Crit Care. 2021;25(1):356. doi:10.1186/s13054-021-03775-3

32. Vasudevan SO, Behl B, Rathinam VA. Pyroptosis-induced inflammation and tissue damage. Semin Immunol. 2023;69:101781. doi:10.1016/j.smim.2023.101781

33. Zhang H, Liu J, Zhou Y, et al. Neutrophil extracellular traps mediate m(6)A modification and regulates sepsis-associated acute lung injury by activating ferroptosis in alveolar epithelial cells. Int J Biol Sci. 2022;18(8):3337–3357. doi:10.7150/ijbs.69141

34. Pang G, Hu K, Ji J, et al. Investigating hub genes in the relationship between septic cardiomyopathy and cuproptosis and potential Chinese herbal drug candidates with bioinformatic tools. Minerva Cardiol Angiol. 2024;72(5):453–464. doi:10.23736/S2724-5683.23.06476-1

35. Yan J, Li Z, Li Y, Zhang Y. Sepsis induced cardiotoxicity by promoting cardiomyocyte cuproptosis. Biochem Biophys Res Commun. 2024;690:149245.

36. Yang J, Chen N, Zhao P, et al. Diminished expression of GLS IN Cd4 + T cells serves as a prognostic indicator associated with cuproptosis in septic patients. Shock. 2024;62(1):51–62. doi:10.1097/SHK.0000000000002370

37. Zhao J, Zhang M, Wang Y, He F, Zhang Q. Identification of cuproptosis-related genes in septic shock based on bioinformatic analysis. PLoS One. 2024;19(12):e0315219. doi:10.1371/journal.pone.0315219

38. Chen X, Kang R, Kroemer G, Tang D. Ferroptosis in infection, inflammation, and immunity. J Exp Med. 2021;218(6). doi:10.1084/jem.20210518

39. Li N, Wang W, Zhou H, et al. Ferritinophagy-mediated ferroptosis is involved in sepsis-induced cardiac injury. Free Radic Biol Med. 2020;160:303–318. doi:10.1016/j.freeradbiomed.2020.08.009

40. Liu P, Feng Y, Li H, et al. Ferrostatin-1 alleviates lipopolysaccharide-induced acute lung injury via inhibiting ferroptosis. Cell Mol Biol Lett. 2020;25(19):10. doi:10.1186/s11658-020-00205-0

41. Xl L, Gy Z, N C. Ferroptosis in sepsis: the mechanism, the role and the therapeutic potential. Front Immunol. 2022;13:956361. doi:10.3389/fimmu.2022.956361

42. Qiu P, Liu Y, Zhang J. Review: the role and mechanisms of macrophage autophagy in sepsis. Inflammation. 2019;42(1):6–19. doi:10.1007/s10753-018-0890-8

43. Saitoh T, Fujita N, Jang MH, et al. Loss of the autophagy protein Atg16L1 enhances endotoxin-induced IL-1beta production. Nature. 2008;456(7219):264–268.

44. Deretic V, Saitoh T, Akira S. Autophagy in infection, inflammation and immunity. Nat Rev Immunol. 2013;13(10):722–737. doi:10.1038/nri3532

45. Unuma K, Aki T, Funakoshi T, Hashimoto K, Uemura K. Extrusion of mitochondrial contents from lipopolysaccharide-stimulated cells: involvement of autophagy. Autophagy. 2015;11(9):1520–1536. doi:10.1080/15548627.2015.1063765

46. Green DR, Levine B. To be or not to be? How selective autophagy and cell death govern cell fate. Cell. 2014;157(1):65–75. doi:10.1016/j.cell.2014.02.049

47. Chen HR, Chuang YC, Chao CH, Yeh TM. Macrophage migration inhibitory factor induces vascular leakage via autophagy. Biol Open. 2015;4(2):244–252. doi:10.1242/bio.201410322

48. Malireddi RKS, Kesavardhana S, Kanneganti TD. ZBP1 and TAK1: master regulators of NLRP3 inflammasome/pyroptosis, apoptosis, and necroptosis (PAN-optosis). Front Cell Infect Microbiol. 2019;9:406. doi:10.3389/fcimb.2019.00406

49. Sun X, Yang Y, Meng X, Li J, Liu X, Liu H. PANoptosis: mechanisms, biology, and role in disease. Immunol Rev. 2024;321(1):246–262. doi:10.1111/imr.13279

50. Karki R, Lee S, Mall R, et al. ZBP1-dependent inflammatory cell death, PANoptosis, and cytokine storm disrupt IFN therapeutic efficacy during coronavirus infection. Science Immunol. 2022;7(74):eabo6294. doi:10.1126/sciimmunol.abo6294

51. Liu J, Hong M, Li Y, Chen D, Wu Y, Hu Y. Programmed cell death tunes tumor immunity. Front Immunol. 2022;13:847345. doi:10.3389/fimmu.2022.847345

52. Zhu P, Ke ZR, Chen JX, Li SJ, Ma TL, Fan XL. Advances in mechanism and regulation of PANoptosis: prospects in disease treatment. Front Immunol. 2023;14:1120034. doi:10.3389/fimmu.2023.1120034

53. Zhou D, Mei Y, Song C, et al. Exploration of the mode of death and potential death mechanisms of nucleus pulposus cells. Eur J Clin Invest. 2024;54(9):e14226. doi:10.1111/eci.14226

54. Karki R, Sharma BR, Tuladhar S, et al. Synergism of TNF-alpha and IFN-gamma triggers inflammatory cell death, tissue damage, and mortality in SARS-CoV-2 infection and cytokine shock syndromes. Cell. 2021;184(1):149–168e117. doi:10.1016/j.cell.2020.11.025

55. Wang Y, Kanneganti TD. From pyroptosis, apoptosis and necroptosis to PANoptosis: a mechanistic compendium of programmed cell death pathways. Comput Struct Biotechnol J. 2021;19:4641–4657. doi:10.1016/j.csbj.2021.07.038

56. Place DE, Lee S, Kanneganti TD. PANoptosis in microbial infection. Curr Opin Microbiol. 2021;59:42–49. doi:10.1016/j.mib.2020.07.012

57. Ocansey DKW, Qian F, Cai P, et al. Current evidence and therapeutic implication of PANoptosis in cancer. Theranostics. 2024;14(2):640–661. doi:10.7150/thno.91814

58. Samir P, Malireddi RKS, Kanneganti TD. The PANoptosome: a deadly protein complex driving pyroptosis, apoptosis, and necroptosis (PANoptosis). Front Cell Infect Microbiol. 2020;10:238. doi:10.3389/fcimb.2020.00238

59. You YP, Yan L, Ke HY, et al. Baicalin inhibits PANoptosis by blocking mitochondrial Z-DNA formation and ZBP1-PANoptosome assembly in macrophages. Acta Pharmacol Sin. 2024;42(2):430–447. doi:10.1038/s41401-024-01376-8

60. Karki R, Kanneganti TD. PANoptosome signaling and therapeutic implications in infection: central role for ZBP1 to activate the inflammasome and PANoptosis. Curr Opin Immunol. 2023;83:102348. doi:10.1016/j.coi.2023.102348

61. Bynigeri RR, Malireddi RKS, Mall R, Connelly JP, Pruett-Miller SM, Kanneganti TD. The protein phosphatase PP6 promotes RIPK1-dependent PANoptosis. BMC Biol. 2024;22(1):122. doi:10.1186/s12915-024-01901-5

62. Sundaram B, Pandian N, Mall R, et al. NLRP12-PANoptosome activates PANoptosis and pathology in response to heme and PAMPs. Cell. 2023;186(13):2783–2801e2720. doi:10.1016/j.cell.2023.05.005

63. Qi Z, Zhu L, Wang K, Wang N. PANoptosis: emerging mechanisms and disease implications. Life Sci. 2023;333:122158. doi:10.1016/j.lfs.2023.122158

64. Lee S, Karki R, Wang Y, Nguyen LN, Kalathur RC, Kanneganti TD. AIM2 forms a complex with pyrin and ZBP1 to drive PANoptosis and host defence. Nature. 2021;597(7876):415–419.

65. Oh S, Lee S. Recent advances in ZBP1-derived PANoptosis against viral infections. Front Immunol. 2023;14:1148727.

66. Asano T, Boisson B, Onodi F, et al. X-linked recessive TLR7 deficiency in ~1% of men under 60 years old with life-threatening COVID-19. Science Immunol. 2021;6(62). doi:10.1126/sciimmunol.abl4348.

67. Channappanavar R, Fehr AR, Zheng J, et al. IFN-I response timing relative to virus replication determines MERS coronavirus infection outcomes. J Clin Invest. 2019;129(9):3625–3639.

68. Moreno-Eutimio MA, Lopez-Macias C, Pastelin-Palacios R. Bioinformatic analysis and identification of single-stranded RNA sequences recognized by TLR7/8 in the SARS-CoV-2, SARS-CoV, and MERS-CoV genomes. Microbes Infect. 2020;22(4–5):226–229.

69. Sumpter R Jr, Loo YM, Foy E, et al. Regulating intracellular antiviral defense and permissiveness to hepatitis C virus RNA replication through a cellular RNA helicase, RIG-I. J Virol. 2005;79(5):2689–2699. doi:10.1128/JVI.79.5.2689-2699.2005

70. Rehwinkel J, Tan CP, Goubau D, et al. RIG-I detects viral genomic RNA during negative-strand RNA virus infection. Cell. 2010;140(3):397–408. doi:10.1016/j.cell.2010.01.020

71. Messaoud-Nacer Y, Culerier E, Rose S, et al. STING agonist diABZI induces PANoptosis and DNA mediated acute respiratory distress syndrome (ARDS). Cell Death Dis. 2022;13(3):269. doi:10.1038/s41419-022-04664-5

72. Malireddi RKS, Gurung P, Mavuluri J, et al. TAK1 restricts spontaneous NLRP3 activation and cell death to control myeloid proliferation. J Exp Med. 2018;215(4):1023–1034. doi:10.1084/jem.20171922

73. Orning P, Weng D, Starheim K, et al. Pathogen blockade of TAK1 triggers caspase-8-dependent cleavage of gasdermin D and cell death. Science. 2018;362(6418):1064–1069. doi:10.1126/science.aau2818

74. Cruz CM, Rinna A, Forman HJ, Ventura AL, Persechini PM, Ojcius DM. ATP activates a reactive oxygen species-dependent oxidative stress response and secretion of proinflammatory cytokines in macrophages. J Biol Chem. 2007;282(5):2871–2879. doi:10.1074/jbc.M608083200

75. Mishra SR, Mahapatra KK, Behera BP, et al. Mitochondrial dysfunction as a driver of NLRP3 inflammasome activation and its modulation through mitophagy for potential therapeutics. Int J Biochem Cell Biol. 2021;136:106013. doi:10.1016/j.biocel.2021.106013

76. Zheng M, Kanneganti TD. The regulation of the ZBP1-NLRP3 inflammasome and its implications in pyroptosis, apoptosis, and necroptosis (PANoptosis). Immunol Rev. 2020;297(1):26–38. doi:10.1111/imr.12909

77. Lee E, Song CH, Bae SJ, Ha KT, Karki R. Regulated cell death pathways and their roles in homeostasis, infection, inflammation, and tumorigenesis. Exp Mol Med. 2023;55(8):1632–1643. doi:10.1038/s12276-023-01069-y

78. Sun L, Wang H, Wang Z, et al. Mixed lineage kinase domain-like protein mediates necrosis signaling downstream of RIP3 kinase. Cell. 2012;148(1–2):213–227. doi:10.1016/j.cell.2011.11.031

79. Faria SS, Costantini S, De lima VCC, et al. NLRP3 inflammasome-mediated cytokine production and pyroptosis cell death in breast cancer. J Biomed Sci. 2021;28(1):26. doi:10.1186/s12929-021-00724-8

80. Shi J, Zhao Y, Wang K, et al. Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death. Nature. 2015;526(7575):660–665. doi:10.1038/nature15514

81. Wang L, Zhu Y, Zhang L, et al. Mechanisms of PANoptosis and relevant small-molecule compounds for fighting diseases. Cell Death Dis. 2023;14(12):851. doi:10.1038/s41419-023-06370-2

82. Dai W, Zheng P, Wu J, et al. Integrated analysis of single-cell RNA-seq and chipset data unravels PANoptosis-related genes in sepsis. Front Immunol. 2023;14:1247131. doi:10.3389/fimmu.2023.1247131

83. Xu J, Zhu M, Luo P, Gong Y. Machine learning screening and validation of PANoptosis-related gene signatures in sepsis. J Inflamm Res. 2024;17:4765–4780. doi:10.2147/JIR.S461809

84. Wang J, Chen S, Chen L, Zhou D. Data-driven analysis that integrates bioinformatics and machine learning uncovers PANoptosis-related diagnostic genes in early pediatric septic shock. Heliyon. 2024;10(18).

85. Lu ZH, Tang Y, Chen H, et al. Identification and functional analysis of PANoptosis-associated genes in the progression from sepsis to ARDS. Immun Inflamm Dis. 2025;13(1):e70136. doi:10.1002/iid3.70136

86. Yang Z, Kao X, Huang N, Yuan K, Chen J, He M. Identification and analysis of PANoptosis-related genes in sepsis-induced lung injury by bioinformatics and experimental verification. J Inflamm Res. 2024;17:1941–1956. doi:10.2147/JIR.S452608

87. Wang Y, Shi Y, Shao Y, Lu X, Zhang H, Miao C. S100A8/A9(hi) neutrophils induce mitochondrial dysfunction and PANoptosis in endothelial cells via mitochondrial complex I deficiency during sepsis. Cell Death Dis. 2024;15(6):462. doi:10.1038/s41419-024-06849-6

88. Gong T, Wang QD, Loughran PA, et al. Mechanism of lactic acidemia-promoted pulmonary endothelial cells death in sepsis: role for CIRP-ZBP1-PANoptosis pathway. Mil Med Res. 2024;11(1):71. doi:10.1186/s40779-024-00574-z

89. Zhang YT, Li HH, Teng F. Inhibition of Piezo1 ameliorates septic cardiomyopathy by blocking calcium-dependent PANoptosis. Eur J Pharmacol. 2025;996:177438. doi:10.1016/j.ejphar.2025.177438

90. Yang Z, Kao X, Zhang L, Huang N, Chen J, He M. Exploring the anti-PANoptosis mechanism of dachaihu decoction against sepsis-induced acute lung injury: network pharmacology, bioinformatics, and experimental validation. Drug Des Devel Ther. 2025;19:349–368. doi:10.2147/DDDT.S495225

91. Liu X, Li Y, Zhang W, et al. Inhibition of cIAP1/2 reduces RIPK1 phosphorylation in pulmonary endothelial cells and alleviate sepsis-induced lung injury and inflammatory response. Immunol Res. 2024;72(4):841–850. doi:10.1007/s12026-024-09491-8

92. Zhou X, Yu X, Wan C, et al. NINJ1 regulates platelet activation and PANoptosis in septic disseminated intravascular coagulation. Int J Mol Sci. 2023;24(4).

93. Zhou X, Xin G, Wan C, et al. Myricetin reduces platelet PANoptosis in sepsis to delay disseminated intravascular coagulation. Biochem Biophys Res Commun. 2024;724:150140. doi:10.1016/j.bbrc.2024.150140

94. Jiang Y, Gao S, Chen Z, et al. Pyroptosis in septic lung injury: interactions with other types of cell death. Biomed Pharmacother. 2023;169:115914. doi:10.1016/j.biopha.2023.115914

95. Cui Y, Wang X, Lin F, et al. MiR-29a-3p improves acute lung injury by reducing alveolar epithelial cell PANoptosis. Aging Dis. 2022;13(3):899–909. doi:10.14336/AD.2021.1023

96. Zhang M, Shang L, Zhou F, et al. Dachengqi decoction dispensing granule ameliorates LPS-induced acute lung injury by inhibiting PANoptosis in vivo and in vitro. J Ethnopharmacol. 2025;336:118699. doi:10.1016/j.jep.2024.118699

97. Zhou R, Ying J, Qiu X, et al. A new cell death program regulated by toll-like receptor 9 through p38 mitogen-activated protein kinase signaling pathway in a neonatal rat model with sepsis associated encephalopathy. Chin Med J. 2022;135(12):1474–1485. doi:10.1097/CM9.0000000000002010

98. Wei S, Wu L, Xiang Z, et al. EIF2AK2 protein targeted activation of AIM2-mediated PANoptosis promotes sepsis-induced acute kidney injury. Renal Failure. 2024;46(2):2403649. doi:10.1080/0886022X.2024.2403649

99. Guo J, Luo Y, Zuo J, Teng J, Shen B, Liu X. Echinacea polyphenols inhibit NLRP3-dependent pyroptosis, apoptosis, and necroptosis via suppressing no production during lipopolysaccharide-induced acute lung injury. J Agric Food Chem. 2023;71(19):7289–7298. doi:10.1021/acs.jafc.2c08382

100. He YQ, Deng JL, Zhou CC, et al. Ursodeoxycholic acid alleviates sepsis-induced lung injury by blocking PANoptosis via STING pathway. Int Immunopharmacol. 2023;125(Pt B):111161.

101. Wang Y, Fu X, Shang Z, et al. In vivo and in vitro study on the regulatory mechanism of XiaoChaiHu decoction on PANoptosis in sepsis-induced cardiomyopathy. J Ethnopharmacol. 2025;336:118740. doi:10.1016/j.jep.2024.118740

102. Oh S, Lee J, Oh J, et al. Integrated NLRP3, AIM2, NLRC4, Pyrin inflammasome activation and assembly drive PANoptosis. Cell Mol Immunol. 2023;20(12):1513–1526. doi:10.1038/s41423-023-01107-9

103. Vande Walle L, Lamkanfi M. Pyroptosis. Curr Biol. 2016;26(13):R568–R572. doi:10.1016/j.cub.2016.02.019

104. Weinlich R, Oberst A, Beere HM, Green DR. Necroptosis in development, inflammation and disease. Nat Rev Mol Cell Biol. 2017;18(2):127–136. doi:10.1038/nrm.2016.149

105. Fritsch M, Gunther SD, Schwarzer R, et al. Caspase-8 is the molecular switch for apoptosis, necroptosis and pyroptosis. Nature. 2019;575(7784):683–687. doi:10.1038/s41586-019-1770-6

106. Liu D, Huang SY, Sun JH, et al. Sepsis-induced immunosuppression: mechanisms, diagnosis and current treatment options. Mil Med Res. 2022;9(1):56. doi:10.1186/s40779-022-00422-y

107. Fajgenbaum DC, June CH. Cytokine Storm. New Engl J Med. 2020;383(23):2255–2273. doi:10.1056/NEJMra2026131

108. Joshi MB, Lad A, Bharath Prasad AS, Balakrishnan A, Ramachandra L, Satyamoorthy K. High glucose modulates IL-6 mediated immune homeostasis through impeding neutrophil extracellular trap formation. FEBS Lett. 2013;587(14):2241–2246. doi:10.1016/j.febslet.2013.05.053

109. Papayannopoulos V. Neutrophil extracellular traps in immunity and disease. Nat Rev Immunol. 2018;18(2):134–147. doi:10.1038/nri.2017.105

110. Muendlein HI, Connolly WM, Magri Z, et al. ZBP1 promotes inflammatory responses downstream of TLR3/TLR4 via timely delivery of RIPK1 to TRIF. Proc Natl Acad Sci USA. 2022;119(24):e2113872119. doi:10.1073/pnas.2113872119

111. Shi FL, Li Q, Xu R, et al. Blocking reverse electron transfer-mediated mitochondrial DNA oxidation rescues cells from PANoptosis. Acta Pharmacol Sin. 2024;45(3):594–608. doi:10.1038/s41401-023-01182-8

112. Pan H, Pan J, Li P, Gao J. Characterization of PANoptosis patterns predicts survival and immunotherapy response in gastric cancer. Clin Immunol. 2022;238:109019. doi:10.1016/j.clim.2022.109019

113. Li H, Boulougoura A, Endo Y, Tsokos GC. Abnormalities of T cells in systemic lupus erythematosus: new insights in pathogenesis and therapeutic strategies. J Autoimmun. 2022;132:102870. doi:10.1016/j.jaut.2022.102870

114. Sun W, Li P, Wang M, et al. Molecular characterization of PANoptosis-related genes with features of immune dysregulation in systemic lupus erythematosus. Clin Immunol. 2023;253:109660. doi:10.1016/j.clim.2023.109660

115. Yang D, Wang X, Sun Y, Shao Y, Shi X. Identification and experimental validation of genes associated with programmed cell death in dendritic cells of the thyroid tissue in Hashimoto’s thyroiditis. Int Immunopharmacol. 2024;142(Pt A):113083. doi:10.1016/j.intimp.2024.113083

116. Zhang X, Tang B, Luo J, et al. Cuproptosis, ferroptosis and PANoptosis in tumor immune microenvironment remodeling and immunotherapy: culprits or new hope. Mol Cancer. 2024;23(1):255. doi:10.1186/s12943-024-02130-8

117. Zheng LY, Duan Y, He PY, et al. Dysregulated dendritic cells in sepsis: functional impairment and regulated cell death. Cell Mol Biol Lett. 2024;29(1):81. doi:10.1186/s11658-024-00602-9

118. van der Poll T, Shankar-Hari M, Wiersinga WJ. The immunology of sepsis. Immunity. 2021;54(11):2450–2464. doi:10.1016/j.immuni.2021.10.012

119. Dong X, Tu H, Bai X, Qin S, Li Z. Intrinsic/extrinsic apoptosis and pyroptosis contribute to the selective depletion of B cell subsets in septic shock patients. Shock. 2023;60(3):345–353. doi:10.1097/SHK.0000000000002174

120. Zhang W, Zhu C, Liao Y, Zhou M, Xu W, Zou Z. Caspase-8 in inflammatory diseases: a potential therapeutic target. Cell Mol Biol Lett. 2024;29(1):130. doi:10.1186/s11658-024-00646-x

121. Moltrasio C, Romagnuolo M, Marzano AV. NLRP3 inflammasome and NLRP3-related autoinflammatory diseases: from cryopyrin function to targeted therapies. Front Immunol. 2022;13:1007705. doi:10.3389/fimmu.2022.1007705

122. Yuan T, Yang HY, Li YP, et al. Scutellarin inhibits inflammatory PANoptosis by diminishing mitochondrial ROS generation and blocking PANoptosome formation. Int Immunopharmacol. 2024;139:112710. doi:10.1016/j.intimp.2024.112710

123. Yang S, Hu C, Chen X, et al. Crosstalk between metabolism and cell death in tumorigenesis. Mol Cancer. 2024;23(1):71. doi:10.1186/s12943-024-01977-1

124. Yang L, Chu Z, Liu M, et al. Amino acid metabolism in immune cells: essential regulators of the effector functions, and promising opportunities to enhance cancer immunotherapy. J Hematol Oncol. 2023;16(1):59. doi:10.1186/s13045-023-01453-1

125. Karki R, Kanneganti TD. The ‘cytokine storm’: molecular mechanisms and therapeutic prospects. Trends Immunol. 2021;42(8):681–705. doi:10.1016/j.it.2021.06.001

126. Yan WT, Zhao WJ, Hu XM, et al. PANoptosis-like cell death in ischemia/reperfusion injury of retinal neurons. Neural Regen Res. 2023;18(2):357–363. doi:10.4103/1673-5374.346545

127. Shi K, Zhang J, Zhou E, Wang J, Wang Y. Small-molecule receptor-interacting protein 1 (RIP1) inhibitors as therapeutic agents for multifaceted diseases: current medicinal chemistry insights and emerging opportunities. J Med Chem. 2022;65(22):14971–14999. doi:10.1021/acs.jmedchem.2c01518

128. Choi S, Keys H, Staples RJ, Yuan J, Degterev A, Cuny GD. Optimization of tricyclic Nec-3 necroptosis inhibitors for in vitro liver microsomal stability. Bioorg Med Chem Lett. 2012;22(17):5685–5688. doi:10.1016/j.bmcl.2012.06.098

129. Qiao S, Zhao WJ, Li HQ, et al. Necrostatin-1 analog DIMO exerts cardioprotective effect against ischemia reperfusion injury by suppressing necroptosis via autophagic pathway in rats. Pharmacology. 2021;106(3–4):189–201. doi:10.1159/000510864

130. Wu Z, Li Y, Cai Y, Yuan J, Yuan C. A novel necroptosis inhibitor-necrostatin-21 and its SAR study. Bioorg Med Chem Lett. 2013;23(17):4903–4906. doi:10.1016/j.bmcl.2013.06.073

131. Brito H, Marques V, Afonso MB, et al. Phenotypic high-throughput screening platform identifies novel chemotypes for necroptosis inhibition. Cell Death Discov. 2020;6(1):6. doi:10.1038/s41420-020-0240-0

132. Martens S, Goossens V, Devisscher L, et al. RIPK1-dependent cell death: a novel target of the Aurora kinase inhibitor Tozasertib (VX-680). Cell Death Dis. 2018;9(2):211. doi:10.1038/s41419-017-0245-7

133. Rui C, Shi SN, Ren W, et al. The multitargeted kinase inhibitor KW-2449 ameliorates cisplatin-induced nephrotoxicity by targeting RIPK1-mediated necroptosis. Biochem Pharmacol. 2021;188:114542. doi:10.1016/j.bcp.2021.114542

134. Xia C, Yao Z, Xu L, Zhang W, Chen H, Zhuang C. Structure-based bioisosterism design of thio-benzoxazepinones as novel necroptosis inhibitors. Eur J Med Chem. 2021;220:113484. doi:10.1016/j.ejmech.2021.113484

135. Li S, Qu L, Wang X, Kong L. Novel insights into RIPK1 as a promising target for future Alzheimer’s disease treatment. Pharmacol Ther. 2022;231:107979. doi:10.1016/j.pharmthera.2021.107979

136. Harris PA, King BW, Bandyopadhyay D, et al. DNA-encoded library screening identifies benzo[b][1,4]oxazepin-4-ones as highly potent and monoselective receptor interacting protein 1 kinase inhibitors. J Med Chem. 2016;59(5):2163–2178. doi:10.1021/acs.jmedchem.5b01898

137. Qin X, Hu L, Shi SN, et al. The Bcr-Abl inhibitor GNF-7 inhibits necroptosis and ameliorates acute kidney injury by targeting RIPK1 and RIPK3 kinases. Biochem Pharmacol. 2020;177:113947. doi:10.1016/j.bcp.2020.113947

138. Zhang C, Guan Q, Shi H, et al. A novel RIP1/RIP3 dual inhibitor promoted OPC survival and myelination in a rat neonatal white matter injury model with hOPC graft. Stem Cell Res Ther. 2021;12(1):462. doi:10.1186/s13287-021-02532-1

139. Feng Y, Hu C, Cui K, et al. GSK840 alleviates retinal neuronal injury by inhibiting RIPK3/MLKL-mediated RGC necroptosis after ischemia/reperfusion. Invest Ophthalmol Vis Sci. 2023;64(14):42. doi:10.1167/iovs.64.14.42

140. Xu CH, Wang JN, Suo XG, et al. RIPK3 inhibitor-AZD5423 alleviates acute kidney injury by inhibiting necroptosis and inflammation. Int Immunopharmacol. 2022;112:109262. doi:10.1016/j.intimp.2022.109262

141. Silva CMS, Wanderley CWS, Veras FP, et al. Gasdermin D inhibition prevents multiple organ dysfunction during sepsis by blocking NET formation. Blood. 2021;138(25):2702–2713. doi:10.1182/blood.2021011525

142. Rathkey JK, Zhao J, Liu Z, et al. Chemical disruption of the pyroptotic pore-forming protein gasdermin D inhibits inflammatory cell death and sepsis. Science Immunol. 2018;3(26). doi:10.1126/sciimmunol.aat2738.

143. Humphries F, Shmuel-Galia L, Ketelut-Carneiro N, et al. Succination inactivates gasdermin D and blocks pyroptosis. Science. 2020;369(6511):1633–1637. doi:10.1126/science.abb9818

144. Ngai WSC, Yang S, Zeng X, et al. Bioorthogonally activatable base editing for on-demand pyroptosis. J Am Chem Soc. 2022;144(12):5411–5417. doi:10.1021/jacs.1c12924

145. Lopez-Pastrana J, Ferrer LM, Li YF, et al. Inhibition of caspase-1 activation in endothelial cells improves angiogenesis: a novel therapeutic potential for ischemia. J Biol Chem. 2015;290(28):17485–17494. doi:10.1074/jbc.M115.641191

146. Wang L, Dong X, Feng S, et al. VX765 alleviates dextran sulfate sodium-induced colitis in mice by suppressing caspase-1-mediated pyroptosis. Int Immunopharmacol. 2022;102:108405. doi:10.1016/j.intimp.2021.108405

147. Xu X, Lan X, Fu S, et al. Dickkopf-1 exerts protective effects by inhibiting PANoptosis and retinal neovascularization in diabetic retinopathy. Biochem Biophys Res Commun. 2022;617(Pt 2):69–76. doi:10.1016/j.bbrc.2022.05.001

148. Ye D, Xu Y, Shi Y, et al. Anti-PANoptosis is involved in neuroprotective effects of melatonin in acute ocular hypertension model. J Pineal Res. 2022;73(4):e12828. doi:10.1111/jpi.12828

149. Ren L, Yang Y, Li W, et al. CDK1 serves as a therapeutic target of adrenocortical carcinoma via regulating epithelial-mesenchymal transition, G2/M phase transition, and PANoptosis. J Transl Med. 2022;20(1):444. doi:10.1186/s12967-022-03641-y

150. Cui B, Qi Z, Liu W, Zhang G, Lin D. ZBP1-mediated PANoptosis: a possible novel mechanism underlying the therapeutic effects of penehyclidine hydrochloride on myocardial ischemia-reperfusion injury. Int Immunopharmacol. 2024;137:112373. doi:10.1016/j.intimp.2024.112373

151. Marchetti C, Swartzwelter B, Gamboni F, et al. OLT1177, a beta-sulfonyl nitrile compound, safe in humans, inhibits the NLRP3 inflammasome and reverses the metabolic cost of inflammation. Proc Natl Acad Sci USA. 2018;115(7):E1530–E1539. doi:10.1073/pnas.1716095115

152. Bakhshi S, Shamsi S. MCC950 in the treatment of NLRP3-mediated inflammatory diseases: latest evidence and therapeutic outcomes. Int Immunopharmacol. 2022;106:108595. doi:10.1016/j.intimp.2022.108595

153. Coll RC, Robertson AA, Chae JJ, et al. A small-molecule inhibitor of the NLRP3 inflammasome for the treatment of inflammatory diseases. Nat Med. 2015;21(3):248–255. doi:10.1038/nm.3806

154. Youm YH, Nguyen KY, Grant RW, et al. The ketone metabolite beta-hydroxybutyrate blocks NLRP3 inflammasome-mediated inflammatory disease. Nat Med. 2015;21(3):263–269. doi:10.1038/nm.3804

155. Huang Y, Jiang H, Chen Y, et al. Tranilast directly targets NLRP3 to treat inflammasome-driven diseases. EMBO Mol Med. 2018;10(4). doi:10.15252/emmm.201708689.

156. Uysal E, Dokur M, Kucukdurmaz F, et al. Targeting the PANoptosome with 3,4-methylenedioxy-β-nitrostyrene, reduces PANoptosis and protects the kidney against renal ischemia-reperfusion injury. J Invest Surg. 2022;35(11–12):1824–1835. doi:10.1080/08941939.2022.2128117

157. Jiao Y, Nan J, Mu B, et al. Discovery of a novel and potent inhibitor with differential species-specific effects against NLRP3 and AIM2 inflammasome-dependent pyroptosis. Eur J Med Chem. 2022;232:114194. doi:10.1016/j.ejmech.2022.114194

158. Kim J, Ahn H, Han BC, et al. Obovatol inhibits NLRP3, AIM2, and non-canonical inflammasome activation. Phytomedicine. 2019;63:153019. doi:10.1016/j.phymed.2019.153019

159. Karki R, Sundaram B, Sharma BR, et al. ADAR1 restricts ZBP1-mediated immune response and PANoptosis to promote tumorigenesis. Cell Rep. 2021;37(3):109858. doi:10.1016/j.celrep.2021.109858

160. Eguchi A, Koyama Y, Wree A, et al. Emricasan, a pan-caspase inhibitor, improves survival and portal hypertension in a murine model of common bile-duct ligation. J Mol Med. 2018;96(6):575–583. doi:10.1007/s00109-018-1642-9

161. Lekakis V, Cholongitas E. The impact of emricasan on chronic liver diseases: current data. Clin J Gastroenterol. 2022;15(2):271–285. doi:10.1007/s12328-021-01585-2

162. Kalil AC, Patterson TF, Mehta AK, et al. Baricitinib plus remdesivir for hospitalized adults with covid-19. New Engl J Med. 2021;384(9):795–807. doi:10.1056/NEJMoa2031994

163. Karki R, Kanneganti TD. Innate immunity, cytokine storm, and inflammatory cell death in COVID-19. J Transl Med. 2022;20(1):542. doi:10.1186/s12967-022-03767-z

164. Zhu Y, Li J, Dai L, Feng W. Ginsenoside Rh2 alleviate sepsis-related encephalopathy via up-regulating Nrf2/HO-1 pathway and apoptosis inhibition. Cell Biochem Biophys. 2024;83(1):587–597. doi:10.1007/s12013-024-01488-2

165. Jiang J, Sun X, Akther M, et al. Ginsenoside metabolite 20(S)-protopanaxatriol from Panax ginseng attenuates inflammation-mediated NLRP3 inflammasome activation. J Ethnopharmacol. 2020;251:112564. doi:10.1016/j.jep.2020.112564

166. Guo D, Cheng K, Song C, et al. Mechanisms of inhibition of nucleus pulposus cells pyroptosis through SDF1/CXCR4-NFkB-NLRP3 axis in the treatment of intervertebral disc degeneration by Duhuo Jisheng decoction. Int Immunopharmacol. 2023;124(Pt A):110844. doi:10.1016/j.intimp.2023.110844

167. Liang Y, Yan Y, Liu N, Wang J, Fang C. Shengxian decoction improves lung function in rats with bleomycin-induced idiopathic pulmonary fibrosis through the inhibition of PANoptosis. J Ethnopharmacol. 2024;329:118153. doi:10.1016/j.jep.2024.118153

168. Lin SY, Chang CL, Liou KT, et al. The protective role of Achyranthes aspera extract against cisplatin-induced nephrotoxicity by alleviating oxidative stress, inflammation, and PANoptosis. J Ethnopharmacol. 2024;319(Pt 1):117097. doi:10.1016/j.jep.2023.117097

169. Cui Z, Liu T, Wen Y, et al. Oral administration of cranberry-derived exosomes attenuates murine premature ovarian failure in association with changes in the specific gut microbiota and diminution in ovarian granulosa cell PANoptosis. Food Funct. 2024;15(23):11697–11714. doi:10.1039/D4FO03468F

Creative Commons License © 2025 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, 4.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.