Back to Journals » Journal of Inflammation Research » Volume 18

PANoptosis: Cross-Talk Among Apoptosis, Necroptosis, and Pyroptosis in Neurological Disorders

Authors Li P, Gao Y, Tao Z, Mu Z, Du S, Zhao X

Received 4 March 2025

Accepted for publication 29 May 2025

Published 19 June 2025 Volume 2025:18 Pages 8131—8140

DOI https://doi.org/10.2147/JIR.S526158

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 2

Editor who approved publication: Dr Adam Bachstetter



Pengpeng Li,1– 3,* Yangyang Gao,4,* Zhenxing Tao,2,5 Zhenqian Mu,1,2 Shiqing Du,2 Xudong Zhao3

1Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, People’s Republic of China; 2Department of Neurosurgery, Jiangnan University Medical Center, Wuxi, Jiangsu, People’s Republic of China; 3Department of Neurosurgery, The Affiliated Wuxi No. 2 People’s Hospital of Nantong Medical University, Wuxi, Jiangsu, People’s Republic of China; 4Ningxia Medical University, Yinchuan, People’s Republic of China; 5Wuxi Neurosurgical Institute, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, People’s Republic of China

*These authors contributed equally to this work

Correspondence: Xudong Zhao, Email [email protected]

Abstract: Cell death mechanisms play a critical role in organismal development and homeostasis, primarily categorized into energy-dependent programmed cell death (PCD) and energy-independent necrotic cell death. PCD, regulated through various forms such as apoptosis, necroptosis, pyroptosis, ferroptosis, and autophagic cell death, is essential for maintaining tissue stability and eliminating abnormal cells. Dysregulation of PCD is associated with numerous diseases, including cancer and neurodegenerative disorders. Recent studies have revealed extensive crosstalk and coordination among classical cell death pathways, leading to the identification of a novel programmed cell death mode termed PANoptosis. PANoptosis involves the dynamic assembly of the PANoptosome complex, which simultaneously activates apoptosis, pyroptosis, and necroptosis pathways in response to pathogen infection or tissue damage. In neurological diseases, PANoptosis exhibits dual roles: it can eliminate pathogen-infected cells but may also exacerbate neuroinflammation and neuronal death, contributing to the progression of neurodegenerative disorders. This review critically evaluates the molecular mechanisms of PANoptosis, its dual roles in neurological diseases (eg, Alzheimer’s disease, Parkinson’s disease, stroke, and glioma), and potential therapeutic strategies targeting PANoptosis, including small-molecule inhibitors, genome editing, and delivery technologies. By addressing conflicting evidence and outstanding questions, this review aims to provide a comprehensive framework for future research and clinical applications. Future research should focus on elucidating the molecular regulatory networks of PANoptosis, developing specific inhibitors, and advancing clinical applications to provide novel insights into the precise treatment of neurological diseases.

Keywords: PANoptosis, neurodegenerative diseases, PANoptosome, inflammasomes, neuroprotection

Introduction

Traditionally, cell death mechanisms are divided into two types: energy - requiring programmed cell death (PCD) and energy - independent necrotic cell death.1,2 Cell death is crucial for organismal development and homeostasis.3 Cell death is crucial for development and homeostasis. During development, it’s essential to remove extra cells for normal morphogenesis and organ formation.4 In adulthood, it’s vital to clear self - reactive immune cells, cancer cells, and damaged cells to maintain internal balance. Too much or too little cell death in the body can lead to human diseases.5 In neurodegenerative diseases, neurons that should survive to maintain nerve function die.6 In cancer, cells that should be eliminated with cancer - causing mutations are not removed.7 Programmed Cell Death (PCD) is a gene - controlled, orderly cell death process.8 It’s crucial for multicellular organism development, tissue stability maintenance, and removal of abnormal cells.2 Key forms of PCD are apoptosis, necroptosis, pyroptosis, ferroptosis, and autophagic cell death, involving different signaling pathways and regulatory mechanisms, such as the extrinsic death receptor and intrinsic mitochondrial pathways.9 PCD is active throughout an organism’s life: in embryonic development, it regulates cell numbers; in the immune system, it clears infected cells.10 Abnormal PCD regulation is associated with various diseases, including cancer, neurodegenerative, cardiovascular, and metabolic diseases (Table 1).11 Therefore, studying PCD mechanisms and regulation is key to understanding life processes and developing disease treatment strategies.12

Table 1 Targeting PANoptosis: Therapeutic Strategies

Classic cell death patterns like apoptosis, pyroptosis, and necroptosis were long considered independent pathways.13 However, recent studies have shown extensive crosstalk and coordination among these pathways.12 In nervous system diseases, apoptotic, pyroptotic, and necroptotic pathways interact via shared molecules like Caspase-8 and RIPK1, and upstream sensors like ZBP1.14 This forms a new programmed cell death pattern called PANoptosis. It was first proposed by Malireddi et al in 2019.15 Panoptosis occurs when cells assemble a PANoptosome complex in response to pathogen infection or tissue damage.16 This complex activates multiple death pathways, triggering an irreversible inflammatory cascade. This complex forms through the involvement of key molecules in pyroptosis, apoptosis, and necroptosis. It also involves epigenetic regulators like HMGB1. These regulators amplify inflammatory signals, creating a positive feedback loop.17

The nervous system is vulnerable to PANoptosis due to its limited regenerative capacity and high energy demands.18 In neurodegenerative diseases like Alzheimer’s and Parkinson’s, panoptosis activation causes neuron death and worsens inflammation.19 In Alzheimer’s disease, β-amyloid oligomers activate ZBP1-PANoptosome in microglia, causing neuronal mitochondrial ROS bursts and tau hyperphosphorylation, accelerating synapse loss.20 In Parkinson’s disease, α-synuclein fibrils activate the NLRP3/ASC axis to trigger PANoptosis in dopaminergic neurons, inducing a cascade that promotes substantia nigra pars compacta neuron degeneration.21 In strokes, ischemic injury can trigger the RIPK3-MLKL pathway through mTOR-ULK1 autophagy imbalance, worsening blood-brain barrier leakage and neuroinflammation.22

Although PANoptosis mainly has pathological effects in neurological diseases, it can also be protective in some cases. For example, in neural infections, it can remove pathogen - infected cells. This limits pathogen spread. This dual nature highlights the need for precise regulation of PANoptosis.23

This review aims to critically evaluate the molecular mechanisms of PANoptosis, its dual roles in nervous system diseases (clearing pathogens vs driving neurodegeneration), and propose targeted intervention strategies. By addressing conflicting evidence and outstanding questions, this review seeks to provide a comprehensive framework for balancing its dual effects and designing multi-modal interventions for precise neuroprotection.

Molecular Mechanisms of Panoptosis

Dynamic Assembly of the PANoptosome Core Complex

The hub of panoptosis is the multi - signal - adapting platform PANoptosome, whose structure varies depending on the type of stimulus.24 In infection models, after ZBP1 recognizes viral nucleic acids (such as influenza virus), it recruits RIPK3, Caspase - 8, and ASC to form the “ZBP1 - PANoptosome”, which activates Caspase - 3, MLKL, and GSDMD, simultaneously driving three death pathways.25 Specifically, ZBP1 recognizes viral nucleic acids through its Zαβ domain, forming liquid - liquid phase - separated condensates. It further binds to RIPK1 and RIPK3 through its RHIM domain to assemble the PANoptosome complex. This process not only activates the Caspase - 3 - mediated apoptotic pathway but also, through the activation of MLKL and GSDMD, mediates necroptosis and pyroptosis, respectively (Figure 1).16

Figure 1 Molecular Mechanisms of PANoptosome Assembly and Activation in Infection and Neurodegenerative Models.

In neurodegenerative models, Aβ/Tau activates the NLRP3 inflammasome, promoting ASC oligomerization and recruiting Pro - Caspase - 1 and Caspase - 8 to form the “NLRP3 - PANoptosome”, leading to IL - 1β release and mitochondrial apoptosis. The activation of the NLRP3 inflammasome is a key step in the inflammatory response in neurodegenerative diseases. By recruiting ASC and Pro - Caspase - 1, it forms the inflammasome complex, which then activates Caspase - 1, promoting the maturation and release of IL - 1β.26 Meanwhile, the recruitment and activation of Caspase - 8 further mediate the mitochondrial apoptotic pathway, resulting in neuronal death.

Key Molecular Interactions

Caspase - 8 has a dual function of cleaving GSDMD (pyroptosis) and activating Caspase - 3 (apoptosis).27 Under TNFα stimulation, RIPK1 promotes necroptosis through its kinase activity or recruits FADD - Caspase - 8 to trigger apoptosis through its scaffolding function.28 Caspase - 8 can directly cleave GSDMD, causing the release of the N - terminal fragment of GSDMD, which inserts into the cell membrane to form pores, thereby triggering pyroptosis.27 In addition, Caspase - 8 can also activate Caspase - 3, which further cleaves substrates such as GSDME, leading to apoptosis.29

Under TNFα stimulation, RIPK1 can drive necroptosis via its kinase activity. When TNFα binds to TNFR1, RIPK1 moves to complex I, activates NF-κB signaling, and boosts anti-apoptotic gene expression.30 When certain conditions arise, RIPK1 can move to complex II, bind to FADD and Caspase-8, and activate Caspase-8, triggering apoptosis. Additionally, RIPK1 can bind to RIPK3, activate MLKL, and induce necroptosis.31 The Interactions and regulatory mechanisms of key molecules reveal the complexity of programmed cell death. They also offer potential targets for developing new therapeutic strategies.

The Role of PANoptosis in Neurological Disorders

Alzheimer’s Disease (AD)

AD is a common neurodegenerative disease mainly affecting the elderly.32 It is characterized by memory loss, cognitive decline, and changes in behavior and personality, significantly impacting patients’ quality of life.33 The pathological features of AD include the deposition of beta - amyloid (Aβ) plaques and the excessive phosphorylation of tau protein, forming neurofibrillary tangles in the brain. These changes lead to neuronal damage and death.34

In AD, Aβ-induced PANoptosis is a complex process involving multiple molecular mechanisms. Specifically, Aβ oligomers can activate TLR4 on neuron membranes, triggering MyD88-dependent NLRP3 inflammasome assembly and IL-1β release, a marker of pyroptosis.20 Concurrently, Caspase-8 induces mitochondrial apoptosis by cleaving Bid protein.20 Clinical evidence shows that in AD patients, the level of GSDMD-NT fragments (pyroptosis markers) in cerebrospinal fluid correlates positively with phosphorylated Tau protein levels. This suggests a link between PANoptosis activation and AD pathology.35 However, conflicting evidence exists regarding the precise role of PANoptosis in AD progression, particularly in the context of Aβ clearance versus neuroinflammation exacerbation. Further research is needed to clarify these mechanisms.

The NLRP3 inflammasome plays a key role in PANoptosis. When activated, it causes the maturation and secretion of IL - 1β and IL - 18. Also, by promoting inflammation and cell death, it worsens AD neuroinflammation.36 Aβ and Tau can be taken up by microglia.37 They destabilize lysosomes and release cathepsin B.38 This process activates the NLRP3 inflammasome. It boosts IL - 1β secretion, thus worsening the disease.39 Recent studies have made significant progress in developing NLRP3 inflammasome inhibitors. These inhibitors, including MCC950, CY-09, OLT1177, Tranilast, and Oridonin, can directly or indirectly inhibit the activation of the NLRP3 inflammasome. This inhibition reduces inflammation and cell death, thereby improving cognitive function in AD patients.40 Additionally, Some traditional Chinese medicine extracts, such as silybin, Lycium polysaccharide, and glycyrrhizin, can lower the protein and mRNA levels of NLRP3, IL-1β, and IL-18. This reduces neuronal damage and improves cognitive function.41

In summary, PANoptosis in AD involves complex mechanisms with cross-regulation of multiple molecules and signaling pathways. Intervening in NLRP3 inflammasome activation and expression can suppress central neuroinflammation, enhance Aβ clearance, and improve cognitive dysfunction. These findings offer a theoretical basis and potential targets for new therapies.

Parkinson’s Disease (PD)

PD is a common neurodegenerative disorder that mainly affects middle-aged and elderly people.42 It is characterized by movement problems such as tremors, stiffness, slow movement, and balance difficulties.43 The pathological features of PD include the significant loss of dopaminergic neurons in the substantia nigra of the midbrain and the formation of Lewy bodies, which are primarily composed of aggregated alpha-synuclein.44

In PD, PANoptosis, a complex cell death form involving pyroptosis, apoptosis, and necroptosis, has gained attention for its mechanisms. One key pathological feature of PD is the abnormal aggregation of α-synuclein (α-Syn). These aggregates cause mitochondrial ROS bursts, activating the RIPK1/Caspase-8 axis and promoting dopaminergic neuron death.45 In addition, the abnormal aggregation of α-Syn can spread between neurons via a prion-like mechanism, causing more neuronal damage and death. This process involves the release, internalization, and misfolding of α-Syn, propagating pathological changes between neurons.46 The PANoptosome assembly involves multiple proteins, including ZBP1, RIPK1, RIPK3, CASP8, and FADD. These proteins interact to form a multimeric complex, starting the PANoptosis process. In animal models, the RIPK1 inhibitor Nec-1s significantly improves motor dysfunction in MPTP-induced mouse models of PD, showing potential therapeutic value.47 However, the precise role of PANoptosis in PD progression remains controversial, particularly in the context of α-Syn propagation and neuroinflammation. Further studies are needed to address these gaps.

Ischemic Stroke

Ischemic stroke, a common cerebrovascular disease, is caused by blocked or severely narrowed brain blood vessels.48 This reduces cerebral blood flow, leading to brain tissue ischemia, hypoxia, and death.49 It has high incidence, prevalence, recurrence, disability, and mortality rates.50

In ischemic stroke, the inflammatory storm during reperfusion is a key pathological process. In the MCAO model, 1 hour of ischemia significantly increases RIPK3 phosphorylation, inducing necroptosis.51 At 24 hours post - reperfusion, Caspase - 1 activation triggers pyroptosis in microglia, further disrupting the blood - brain barrier.52 During this process, PANoptosis is triggered. It causes massive neuronal and microglial death, worsening brain tissue damage.53 PANoptosis, which encompasses pyroptosis, apoptosis, and necroptosis, plays a key role in ischemic stroke pathophysiology. Researching PANoptosis mechanisms in ischemic stroke can aid new therapy development. This can reduce reperfusion injury and enhance patient recovery. However, the dual role of PANoptosis in ischemic stroke, particularly its protective versus detrimental effects, remains unclear and warrants further investigation.

Hemorrhagic Stroke

Hemorrhagic stroke, an acute cerebrovascular disease caused by cerebral vessel rupture, has high mortality and disability rates.54 It often suddenly occurs during activity, manifesting as headache, nausea, vomiting, impaired consciousness, and limb paralysis.49 Depending on the bleeding site, it is mainly divided into intracerebral hemorrhage and subarachnoid hemorrhage.55

In hemorrhagic stroke, free hemin upregulates HMGB1 via TLR4, triggering microglial PANoptosis and releasing IL-18, which worsens brain edema.56 This process is crucial in the pathophysiology of hemorrhagic stroke. Studies show that after brain hemorrhage, erythrocyte components like hemoglobin and its degradation products (heme and iron) are released.57 The degraded iron ions induce lipid peroxidation, producing free radicals that damage the blood-brain barrier and worsen cerebral edema.58 In addition, TLR4 signaling activation is closely related to neuron apoptosis, and modulating this pathway can mediate neuron apoptosis.59 Studying PANoptosis mechanisms in hemorrhagic stroke can help develop new treatments to reduce brain edema and neuron damage, improving patient outcomes. However, the precise mechanisms underlying PANoptosis in hemorrhagic stroke remain poorly understood, and further research is needed to clarify its role in disease progression.

Glioma

Gliomas, the most common primary tumors in the central nervous system, originate from neural glial cells (such as astrocytes and oligodendrocytes), accounting for about 30% of all brain tumors.60 According to the WHO grading system, gliomas can be classified into grades I - IV, with glioblastoma (GBM, grade IV) being the most malignant, highly invasive, and prone to recurrence, having a median survival period of only 12–18 months.61 Currently, standard treatments for gliomas mainly include surgical resection combined with radiochemotherapy (eg, temozolomide). However, these treatments often face challenges such as drug resistance and difficulty penetrating the blood-brain barrier.62

Panoptosis, by regulating the cleavage of Caspase family proteins (such as CASP1/3/8) and Gasdermin proteins (such as GSDMD), plays multiple roles in gliomas.63 GPX8-mediated panoptosis drives microglia to tumors, releases DAMPs, and boosts tumor immunogenicity. It also reshapes the immune microenvironment by increasing T-cell infiltration and activating dendritic cells, improving the response to immune therapies like PD-1/PD-L1 inhibitors.64 Panoptosis-related genes (eg, ZBP1, NLRP3) are linked to glioma prognosis. They can guide personalized treatments, such as predicting temozolomide resistance or radiotherapy sensitivity.65

Targeting panoptosis (eg, disrupting PANoptosome assembly or Gasdermin function) can reverse chemotherapy resistance in high-grade gliomas.66 It also works with oncolytic viruses and CAR-T therapy to enhance antitumor immune responses. This approach offers new ways to develop low-toxicity drugs and improve combination therapies.67 However, the dual role of PANoptosis in glioma progression, particularly its impact on tumor immunogenicity versus tumor cell survival, remains controversial. Future research should focus on clarifying these mechanisms to optimize therapeutic strategies.

Targeting PANoptosis: Therapeutic Strategies

Small Molecule Inhibitor Development

NLRP3 Inhibitors

NLRP3 inhibitors are compounds designed to target the NLRP3 inflammasome, a critical component of the innate immune system involved in detecting microbial infections and cellular damage. By suppressing NLRP3, these compounds can modulate inflammatory responses, prevent tissue damage, and reduce the release of pro-inflammatory cytokines (eg, IL-1β and IL-18), as well as limit inflammatory necrosis.68

NLRP3 inhibitors show significant therapeutic potential in neurological diseases. In AD models, like APP/PS1 mice, the NLRP3 inhibitor MCC950 demonstrates remarkable therapeutic effects.69 For example, intraperitoneal injection of 10 mg / kg MCC950 daily reduces Aβ plaques by 50% and significantly improves cognitive function in APP/PS1 mice. This may be due to MCC950’s direct inhibition of the NLRP3 inflammasome, reducing inflammation and neuronal damage. Additionally, MCC950 promotes non - inflammatory Aβ clearance, further improving cognitive function.70

Besides Alzheimer’s, NLRP3 inhibitors show promise in other neurological diseases. In ischemia - reperfusion models, MCC950 significantly reduces brain injury and improves neurologic function post - ischemic stroke.71 Additionally, MCC950 exerts neuroprotective effects by modulating intracellular calcium signaling pathways and autophagy.72

Developing NLRP3 inhibitors with novel structures and mechanisms is a hot research area. For instance, Sorbremnoids A and B, found in the fungus Penicillium citrinum, are new NLRP3 inhibitors. They directly target the NLRP3 protein, inhibiting NLRP3 inflammasome assembly and activation, and show potential for treating inflammatory and neurodegenerative diseases. These compounds have significant anti - inflammatory activity in diabetic wound healing models, accelerating healing and suggesting clinical potential.26

In addition, other NLRP3 inhibitors, such as CY-09, OLT1177, Tranilast, and Oridonin, have shown therapeutic potential in various neurological disease models.73–75 These inhibitors exert neuroprotective effects by directly or indirectly inhibiting NLRP3 inflammasome activation, reducing inflammation and cell death.

In summary, NLRP3 inhibitors like MCC950 show significant therapeutic potential in various neurological diseases by inhibiting the NLRP3 inflammasome and reducing neuroinflammation. The ongoing development of new NLRP3 inhibitors with improved efficacy and safety may offer novel treatment options for patients with neurological diseases.

Broad-Spectrum Apoptosis Inhibitors

Broad - spectrum apoptosis inhibitors are substances that can block multiple apoptosis pathways by targeting key apoptotic signaling molecules, thus protecting cells from programmed death.78 These compounds show potential therapeutic value in various disease models, especially in neurodegenerative and cardiovascular diseases.

Broad - spectrum apoptosis inhibitors have promising therapeutic effects in neurological diseases. Emricasan (IDN - 6556), an irreversible pan - caspase inhibitor, targets key apoptotic effector proteins like caspase - 3, - 8, and - 9, reducing neuronal apoptosis.76 In vitro studies show Emricasan has neuroprotective effects in astrocyte models and blocks caspase-3 activation caused by Zika virus.79

Other broad - spectrum apoptosis inhibitors, like Q - VD - OPh and Z - VAD - FMK, have also been widely studied. Q - VD - OPh inhibits caspase - 1, - 3, - 8, and - 9 activities, reducing neuronal death and infarct volume in stroke models.77 Z-VAD-FMK, on the other hand, blocks the pan-caspase pathway to inhibit Iron-overloaded rats, which is a form of axonal degeneration.80 These drugs work by inhibiting mitochondrial apoptosis and death receptor pathways. They also regulate inflammasome activation (eg, NLRP3) and reduce pro - inflammatory cytokine release, such as IL - 1β. This dual action helps alleviate neuroinflammation and glial activation.

Future research must tackle key challenges like enhancing blood - brain - barrier penetration with optimized drug delivery systems (eg, nanoparticles or exosomes) and developing precise biomarkers to identify optimal treatment windows. Also, chronic apoptosis inhibition might disrupt physiological cell clearance and increase cancer risk, so careful evaluation is needed in clinical translation.

Genome Editing and Delivery Technologies

Genome editing tech like CRISPR - Cas9 enables precise genome modifications. CRISPR - Cas9 comprises a Cas9 nuclease and an sgRNA, which identifies and binds specific DNA sequences, induces double - strand breaks, and allows gene editing. Delivery tech is essential for introducing genome editing tools or therapeutic genes into cells, with common methods including viral and non - viral vectors.81

PANoptosis-targeting genome editing and delivery technologies have shown remarkable potential in treating neurological disorders. For example, CRISPR/Cas9 has achieved groundbreaking progress in Huntington’s disease (HD) treatment. In HD pig models, adeno-associated virus (AAV)-delivered CRISPR/Cas9 systems successfully targeted the CAG repeat expansion region in the mutant Huntington gene (HTT) and replaced it with normal sequences. This intervention significantly reduced mutant protein neurotoxicity and improved motor deficits.82 Similar applications are being explored in Alzheimer’s and Parkinson’s diseases, such as targeting the APP or α-synuclein genes to reduce pathological protein aggregation.83

CRISPR interference (CRISPRi), which uses a catalytically inactive Cas9 (dCas9) to inhibit specific gene transcription, offers a novel strategy for modulating neuroinflammation or apoptosis-related pathways.

In neurological gene therapy, viral vectors (eg, AAV) are the top choice due to their efficient blood - brain - barrier penetration and long - term expression. For instance, AAV9 vectors successfully delivered CRISPR components to the brain and achieved precise editing in HD pig models via intracranial or intravenous injection.84 Non - viral delivery systems, like liposomes and exosomes, are also being studied. They have advantages such as low immunogenicity and repeatable administration. However, research is still focused on improving their targeting efficiency and delivery accuracy.

Future research should focus on developing more precise delivery methods, like targeting specific neuron subtypes, and combination therapies, such as combining genome editing with neurotrophic factor delivery, to improve treatment effectiveness and minimize off-target effects. However, long-term safety issues, including immune reactions and cancer-related risks, need careful assessment through large-scale preclinical studies.

Challenges and Future Directions

Research on PANoptosis faces challenges in understanding its complex molecular mechanisms, developing specific inhibitors, and advancing clinical applications. However, ongoing research and technological progress are increasingly revealing its therapeutic potential. Future research should focus on a deeper understanding of PANoptosis molecular mechanisms, particularly the assembly and regulation of PANoptosomes, which could pave the way for the identification of more specific therapeutic targets. Additionally, the development of novel inhibitors targeting PANoptosome sensors and other components of the PANoptosis pathway may yield improved therapeutic outcomes.

Furthermore, using multi-omics technologies to analyze PANoptosis regulatory networks can reveal new molecular targets and biomarkers. Another key direction is advancing clinical trials for PANoptosis-related drugs, particularly for neurodegenerative diseases and cancer. Nanomedicine has great potential in developing new tumor-targeted PANoptosis-inducing agents. Also, integrative approaches, like combining traditional Chinese and Western medicine or applying photodynamic therapy, open up new avenues for PANoptosis research.

In summary, PANoptosis research holds significant promise in elucidating molecular mechanisms, advancing drug development, and expanding clinical applications. Future progress in PANoptosis research should be driven by interdisciplinary collaboration, multi-omics technologies, nanomedicine, and integrative therapies. However, addressing conflicting evidence and outstanding questions, particularly regarding the dual roles of PANoptosis in disease progression, remains a critical challenge that requires further investigation.

Disclosure

The authors declare that they have no competing interests.

References

1. Elmore S. Apoptosis: a review of programmed cell death. Toxicol Pathol. 2007;35(4):495–516. doi:10.1080/01926230701320337

2. Zhang YY, Chen X, Gueydan C, et al. Plasma membrane changes during programmed cell deaths. Cell Res. 2018;28(1):9–21. doi:10.1038/cr.2017.133

3. Newton K, Strasser A, Kayagaki N, et al. Cell death. Cell. 2024;187(2):235–256. doi:10.1016/j.cell.2023.11.044

4. Glover HL, Schreiner A, Dewson G, et al. Mitochondria and cell death. Nat Cell Biol. 2024;26(9):1434–1446. doi:10.1038/s41556-024-01429-4

5. Robinson N, Ganesan R, Hegedűs C, et al. Programmed necrotic cell death of macrophages: focus on pyroptosis, necroptosis, and parthanatos. Redox Biol. 2019;26:101239. doi:10.1016/j.redox.2019.101239

6. Moujalled D, Strasser A, Liddell JR. Molecular mechanisms of cell death in neurological diseases. Cell Death Differ. 2021;28(7):2029–2044. doi:10.1038/s41418-021-00814-y

7. Yuan JY, Ofengeim D. A guide to cell death pathways. Nat Rev Mol Cell Biol. 2024;25(5):379–395. doi:10.1038/s41580-023-00689-6

8. 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

9. Jorgensen I, Rayamajhi M, Miao EA. Programmed cell death as a defence against infection. Nat Rev Immunol. 2017;17(3):151–164. doi:10.1038/nri.2016.147

10. Chen Y, Li X, Yang M, Liu SB. Research progress on morphology and mechanism of programmed cell death. Cell Death Dis. 2024;15(5):327.

11. Bedoui S, Herold MJ, Strasser A. Emerging connectivity of programmed cell death pathways and its physiological implications. Nat Rev Mol Cell Biol. 2020;21(11):678–695. doi:10.1038/s41580-020-0270-8

12. 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). doi:10.1038/s41419-023-06370-2

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

14. Lou JS, Mao Y, Jiang W, et al. TRIM56 modulates YBX1 degradation to ameliorate ZBP1-mediated neuronal PANoptosis in spinal cord injury. Adv Sci. 2024;11(42). doi:10.1002/advs.202407132

15. 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. doi:10.3389/fcimb.2019.00406

16. 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

17. Kwak MS, Choi S, Kim J, et al. SARS-CoV-2 infection induces HMGB1 secretion through post-translational modification and PANoptosis. Immun Net. 2023;23(3). doi:10.4110/in.2023.23.e26

18. Sun YY, Zhu CL. Potential role of PANoptosis in neuronal cell death: commentary on “PANoptosis-like cell death in ischemia/reperfusion injury of retinal neurons”. Neural Regeneration Res. 2023;18(2):339–340. doi:10.4103/1673-5374.346483

19. Song R, Yin S, Wu J, et al. Neuronal regulated cell death in aging-related neurodegenerative diseases: key pathways and therapeutic potentials. Neural Regeneration Res. 2025;20(8):2245–2263. doi:10.4103/NRR.NRR-D-24-00025

20. Meng XY, Song Q, Liu Z, Liu X, Wang Y, Liu J. Neurotoxic β-amyloid oligomers cause mitochondrial dysfunction-the trigger for PANoptosis in neurons. Front Aging Neurosci. 2024;16.

21. Wu KM, Xu Q-H, Liu Y-Q, et al. Neuronal FAM171A2 mediates α-synuclein fibril uptake and drives Parkinson’s disease. Science. 2025;387(6736):892–900. doi:10.1126/science.adp3645

22. Wang Y, Xu M, Zuo X, et al. The critical role of MLKL in hemorrhagic stroke and the therapeutic potential of its associated protein network. Front Cell Develop Biol. 2025;12. doi:10.3389/fcell.2024.1509877

23. 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

24. Karki R, Lee S, Mall R, et al. ZBP1-dependent inflammatory cell death, PANoptosis, and cytokine storm disrupt IFN therapeutic efficacy during coronavirus infection. Sci Immunol. 2022;8(90):eabo6294.

25. Xie FY, Wu D, Huang J, et al. ZBP1 condensate formation synergizes Z-NAs recognition and signal transduction. Cell Death Dis. 2024;15(7). doi:10.1038/s41419-024-06889-y

26. Feng SY, Wierzbowski MC, Hrovat-Schaale K, et al. Mechanisms of NLRP3 activation and inhibition elucidated by functional analysis of disease-associated variants. Nat Immunol. 2025;26(3):511–523. doi:10.1038/s41590-025-02088-9

27. Newton K, Wickliffe KE, Maltzman A, et al. Activity of caspase-8 determines plasticity between cell death pathways. Nature. 2019;575(7784):679–+. doi:10.1038/s41586-019-1752-8

28. Zhang W, Liu H, Zhang D, et al. Role of hepatocyte RIPK1 in maintaining liver homeostasis during metabolic challenges. Elife. 2025;13. doi:10.7554/eLife.96798.3

29. Amusan OT, Wang S, Yin C, et al. RIPK1 is required for ZBP1-driven necroptosis in human cells. PLoS Biol. 2025;23(2):e3002845. doi:10.1371/journal.pbio.3002845

30. Izumi Y, O’Dell KA, Mennerick S, et al. Effects of acute pro-inflammatory stimulation and 25-hydroxycholesterol on hippocampal plasticity and learning involve NLRP3 inflammasome and cellular stress responses. Sci Rep. 2025;15(1):6149. doi:10.1038/s41598-025-90149-2

31. Yu L, Guo Q, Li Y, et al. CHMP4C promotes pancreatic cancer progression by inhibiting necroptosis via the RIPK1/RIPK3/MLKL pathway. J Adv Res. 2025. doi:10.1016/j.jare.2025.01.040

32. DeTure MA, Dickson DW. The neuropathological diagnosis of Alzheimer’s disease. Mol Neurodegeneration. 2019;14(1). doi:10.1186/s13024-019-0333-5

33. Jack CR, Bennett DA, Blennow K, et al. NIA-AA research framework: toward a biological definition of Alzheimer’s disease. Alzheimers Dementia. 2018;14(4):535–562. doi:10.1016/j.jalz.2018.02.018

34. Bai RR, Guo J, Ye X-Y, et al. Oxidative stress: the core pathogenesis and mechanism of Alzheimer’s disease. Ageing Res Rev. 2022;77:101619. doi:10.1016/j.arr.2022.101619

35. Wojdala AL, Bellomo G, Gaetani L, et al. Immunoassay detection of multiphosphorylated tau proteoforms as cerebrospinal fluid and plasma Alzheimer’s disease biomarkers. Nat Commun. 2025;16(1). doi:10.1038/s41467-024-54878-8

36. Pennisi M, Crupi R, Di Paola R, et al. Inflammasomes, hormesis, and antioxidants in neuroinflammation: role of NRLP3 in Alzheimer disease. J Neurosci Res. 2017;95(7):1360–1372. doi:10.1002/jnr.23986

37. Evering TH, Marston JL, Gan L, et al. Transposable elements and Alzheimer’s disease pathogenesis. Trends Neurosci. 2023;46(3):170–172. doi:10.1016/j.tins.2022.12.003

38. Sundelöf J, Sundström J, Hansson O, et al. Higher cathepsin B levels in plasma in Alzheimer’s disease compared to healthy controls. J Alzheimers Dis. 2010;22(4):1223–1230. doi:10.3233/JAD-2010-101023

39. Liu HJ, You M, Yang H, et al. Exploring the molecular characterization of PANoptosis-related genes with features of immune dysregulation in Alzheimer’s disease based on bulk and single-nuclei RNA sequencing. Metab Brain Dis. 2025;40(1). doi:10.1007/s11011-025-01540-x

40. McManus RM, Latz E. NLRP3 inflammasome signalling in Alzheimer’s disease. Neuropharmacology. 2024;252:109941. doi:10.1016/j.neuropharm.2024.109941

41. He JY, Deng Y, Ren L, et al. Isoliquiritigenin from licorice flavonoids attenuates NLRP3-mediated pyroptosis by SIRT6 in vascular endothelial cells. J Ethnopharmacol. 2023;303:115952. doi:10.1016/j.jep.2022.115952

42. Cramb KML, Beccano-Kelly D, Cragg SJ, et al. Impaired dopamine release in Parkinson’s disease. Brain. 2023;146(8):3117–3132. doi:10.1093/brain/awad064

43. Bloem BR, Okun MS, Klein C. Parkinson’s disease. Lancet. 2021;397(10291):2284–2303. doi:10.1016/S0140-6736(21)00218-X

44. Vázquez-Vélez GE, Zoghbi HY. Parkinson’s disease genetics and pathophysiology. In: Roska B, Zoghbi HY, editors. Annual Review of Neuroscience, Vol 44, 2021. 2021:87–108.

45. Xu Q, Wang H, Yang R, et al. α-Synuclein amyloid fibril directly binds to LC3B and suppresses SQSTM1/p62-mediated selective autophagy. Cell Res. 2025;35(1):72–75. doi:10.1038/s41422-024-01022-2

46. Park H, Kam T-I, Dawson VL, et al. α-Synuclein pathology as a target in neurodegenerative diseases. Nat Rev Neurol. 2025;21(1):32–47. doi:10.1038/s41582-024-01043-w

47. Stocchi F, Bravi D, Emmi A, et al. Parkinson disease therapy: current strategies and future research priorities. Nat Rev Neurol. 2024;20(12):695–707. doi:10.1038/s41582-024-01034-x

48. Feske SK. Ischemic Stroke. Am J Med. 2021;134(12):1457–1464. doi:10.1016/j.amjmed.2021.07.027

49. Li P, Tao Z, Zhao X. The role of osteopontin (OPN) in regulating microglia phagocytosis in nervous system diseases. JIN. 2024;23(9). doi:10.31083/j.jin2309169

50. DeLong JH, Ohashi SN, O’Connor KC, et al. Inflammatory responses after ischemic stroke. Semin Immunopathol. 2022;44(5):625–648. doi:10.1007/s00281-022-00943-7

51. Zhang YY, Liu W-N, Li Y-Q, et al. Ligustroflavone reduces necroptosis in rat brain after ischemic stroke through targeting RIPK1/RIPK3/MLKL pathway. Naunyn-Schmiedebergs Arch Pharmacol. 2019;392(9):1085–1095. doi:10.1007/s00210-019-01656-9

52. Tian HY, Lei Y-X, Zhou J-T, et al. Insight into interplay between PANoptosis and autophagy: novel therapeutics in ischemic stroke. Front Mol Neurosci. 2025;17. doi:10.3389/fnmol.2024.1482015

53. Lan ZW, Tan F, He J, et al. Curcumin-primed olfactory mucosa-derived mesenchymal stem cells mitigate cerebral ischemia/reperfusion injury-induced neuronal PANoptosis by modulating microglial polarization. Phytomedicine. 2024;129:155635. doi:10.1016/j.phymed.2024.155635

54. Tu WJ, Wang LD, Yan F; Special Writing Grp China. China stroke surveillance report 2021. Military Med Res. 2023;10(1). doi:10.1186/s40779-023-00463-x

55. Ohashi SN, DeLong JH, Kozberg MG, et al. Role of inflammatory processes in hemorrhagic stroke. Stroke. 2023;54(2):605–619. doi:10.1161/STROKEAHA.122.037155

56. Le K, Chibaatar Daliv E, Wu S, et al. SIRT1-regulated HMGB1 release is partially involved in TLR4 signal transduction: a possible anti-neuroinflammatory mechanism of resveratrol in neonatal hypoxic-ischemic brain injury. Int Immunopharmacol. 2019;75:105779. doi:10.1016/j.intimp.2019.105779

57. Li P, Tao Z, Gao Y, et al. Ability of SPP1 to alleviate post-intracerebral hemorrhage ferroptosis via Nrf2/HO1 pathway. Brain Behav. 2025;15(5):e70493. doi:10.1002/brb3.70493

58. Keep RF, Hua Y, Xi GH. Intracerebral haemorrhage: mechanisms of injury and therapeutic targets. Lancet Neurol. 2012;11(8):720–731. doi:10.1016/S1474-4422(12)70104-7

59. Fang H, Wang P-F, Zhou Y, et al. Toll-like receptor 4 signaling in intracerebral hemorrhage-induced inflammation and injury. J Neuroinflammation. 2013;10(1). doi:10.1186/1742-2094-10-27

60. Ostrom QT, Price M, Neff C, et al. CBTRUS statistical report: primary brain and other central nervous system tumors diagnosed in the United States in 2015-2019. Neuro-Oncology. 2022;24(Supplement_5):v1–v95. doi:10.1093/neuonc/noac202

61. Figarella-Branger D, Appay R, Metais A, et al. The 2021 WHO classification of tumours of the central nervous system. Annales de Pathologie. 2022;42(5):367–382. doi:10.1016/j.annpat.2021.11.005

62. Stupp R, Taillibert S, Kanner A, et al. Effect of tumor-treating fields plus maintenance temozolomide vs maintenance temozolomide alone on survival in patients with glioblastoma A randomized clinical trial. JAMA J Am Med Assoc. 2017;318(23):2306–2316. doi:10.1001/jama.2017.18718

63. Yu D, Wang S, Wang J, et al. EZH2-STAT3 signaling pathway regulates GSDMD-mediated pyroptosis in glioblastoma. Cell Death Discovery. 2024;10(1). doi:10.1038/s41420-024-02105-0

64. Chen ZG, Zheng D, Lin Z, et al. GPX8 regulates pan-apoptosis in gliomas to promote microglial migration and mediate immunotherapy responses. Front Immunol. 2023;14:1260169.

65. Song J, Xu Z, Fan Q, et al. The PANoptosis-related signature indicates the prognosis and tumor immune infiltration features of gliomas. Front Mol Neurosci. 2023;16:1198713. doi:10.3389/fnmol.2023.1198713

66. Gao J, Xiong A, Liu J, et al. PANoptosis: bridging apoptosis, pyroptosis, and necroptosis in cancer progression and treatment. Cancer Genet Ther. 2024;31(7):970–983. doi:10.1038/s41417-024-00765-9

67. Cai Y, Xiao H, Xue S, et al. Integrative analysis of immunogenic PANoptosis and experimental validation of cinobufagin-induced activation to enhance glioma immunotherapy. J Exp Clin Cancer Res. 2025;44(1):35. doi:10.1186/s13046-025-03301-1

68. Ma Q. Pharmacological inhibition of the NLRP3 inflammasome: structure, molecular activation, and inhibitor-NLRP3 interaction. Pharmacol Rev. 2023;75(3):487–520. doi:10.1124/pharmrev.122.000629

69. Dempsey C, Rubio Araiz A, Bryson KJ, et al. Inhibiting the NLRP3 inflammasome with MCC950 promotes non-phlogistic clearance of amyloid-β and cognitive function in APP/PS1 mice. Brain Behav Immun. 2017;61:306–316. doi:10.1016/j.bbi.2016.12.014

70. Py BF. Functional analysis of NLRP3 variants provides insight into inflammasome regulation. Nat Immunol. 2025;26(3):337–339. doi:10.1038/s41590-025-02093-y

71. Wu X, Wang B, Zhou Y, et al. NLRP3 inflammasome inhibitor MCC950 reduces cerebral ischemia/reperfusion induced neuronal ferroptosis. Neurosci Lett. 2023;795:137032. doi:10.1016/j.neulet.2022.137032

72. Naeem A, Prakash R, Kumari N, et al. MCC950 reduces autophagy and improves cognitive function by inhibiting NLRP3-dependent neuroinflammation in a rat model of Alzheimer’s disease. Brain Behav Immun. 2024;116:70–84. doi:10.1016/j.bbi.2023.11.031

73. Fang M, Xia F, Wang J, et al. The NLRP3 inhibitor, OLT1177 attenuates brain injury in experimental intracerebral hemorrhage. Int Immunopharmacol. 2024;131:111869. doi:10.1016/j.intimp.2024.111869

74. Wang Y, Liu Y-J, Zhang -M-M, et al. CY-09 alleviates the depression-like behaviors via inhibiting NLRP3 inflammasome-mediated neuroinflammation in lipopolysaccharide-induced mice. ACS Chem Neurosci. 2022;13(23):3291–3302. doi:10.1021/acschemneuro.2c00348

75. Zhuo Y, Zhuo J. Tranilast treatment attenuates cerebral ischemia-reperfusion injury in rats through the inhibition of inflammatory responses mediated by NF-κB and PPARs. Cts-Clin Transl Sci. 2019;12(2):196–202. doi:10.1111/cts.12606

76. Xu M, Lee EM, Wen Z, et al. Identification of small-molecule inhibitors of Zika virus infection and induced neural cell death via a drug repurposing screen. Nat Med. 2016;22(10):1101–1107. doi:10.1038/nm.4184

77. Teng X, Chen W, Liu Z, et al. NLRP3 inflammasome is involved in Q-VD-OPH induced necroptosis following cerebral ischemia-reperfusion injury. Neurochem Res. 2018;43(6):1200–1209. doi:10.1007/s11064-018-2537-4

78. Mohammad RM, Muqbil I, Lowe L, et al. Broad targeting of resistance to apoptosis in cancer. Semi Cancer Biol. 2015;35:S78–S103. doi:10.1016/j.semcancer.2015.03.001

79. Alam A, Imam N, Farooqui A, et al. Recent trends in ZikV research: a step away from cure. Biomed Pharmacother. 2017;91:1152–1159. doi:10.1016/j.biopha.2017.05.045

80. Sripetchwandee J, Kongkaew A, Kumfu S, et al. Ferrostatin-1 and Z-VAD-FMK potentially attenuated Iron-mediated neurotoxicity and rescued cognitive function in Iron-overloaded rats. Life Sci. 2023;313:121269. doi:10.1016/j.lfs.2022.121269

81. Liu C, Zhang L, Liu H, et al. Delivery strategies of the CRISPR-Cas9 gene-editing system for therapeutic applications. J Control Release. 2017;266:17–26. doi:10.1016/j.jconrel.2017.09.012

82. Lin YQ, Li C, Chen Y, et al. RNA-Targeting CRISPR/CasRx system relieves disease symptoms in Huntington’s disease models. Mol Neurodegeneration. 2025;20(1). doi:10.1186/s13024-024-00794-w

83. Paquet D, Kwart D, Chen A, et al. Efficient introduction of specific homozygous and heterozygous mutations using CRISPR/Cas9. Nature. 2016;533(7601):125–+. doi:10.1038/nature17664

84. Yan S, Zheng X, Lin Y, et al. Cas9-mediated replacement of expanded CAG repeats in a pig model of Huntington’s disease. Nat Biomed Eng. 2023;7(5):629–+. doi:10.1038/s41551-023-01007-3

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.