Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Oncology Letters
      • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Biomedical Reports
      • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • Information for Authors
    • Information for Reviewers
    • Information for Librarians
    • Information for Advertisers
    • Conferences
  • Language Editing
Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • For Authors
    • For Reviewers
    • For Librarians
    • For Advertisers
    • Conferences
  • Language Editing
Login Register Submit
  • This site uses cookies
  • You can change your cookie settings at any time by following the instructions in our Cookie Policy. To find out more, you may read our Privacy Policy.

    I agree
Search articles by DOI, keyword, author or affiliation
Search
Advanced Search
presentation
International Journal of Molecular Medicine
Join Editorial Board Propose a Special Issue
Print ISSN: 1107-3756 Online ISSN: 1791-244X
Journal Cover
December-2025 Volume 56 Issue 6

Full Size Image

Sign up for eToc alerts
Recommend to Library

Journals

International Journal of Molecular Medicine

International Journal of Molecular Medicine

International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.

International Journal of Oncology

International Journal of Oncology

International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.

Molecular Medicine Reports

Molecular Medicine Reports

Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.

Oncology Reports

Oncology Reports

Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.

Oncology Letters

Oncology Letters

Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.

Biomedical Reports

Biomedical Reports

Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.

Molecular and Clinical Oncology

Molecular and Clinical Oncology

International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.

World Academy of Sciences Journal

World Academy of Sciences Journal

Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.

International Journal of Functional Nutrition

International Journal of Functional Nutrition

Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.

International Journal of Epigenetics

International Journal of Epigenetics

Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.

Medicine International

Medicine International

An International Open Access Journal Devoted to General Medicine.

Journal Cover
December-2025 Volume 56 Issue 6

Full Size Image

Sign up for eToc alerts
Recommend to Library

  • Article
  • Citations
    • Cite This Article
    • Download Citation
    • Create Citation Alert
    • Remove Citation Alert
    • Cited By
  • Similar Articles
    • Related Articles (in Spandidos Publications)
    • Similar Articles (Google Scholar)
    • Similar Articles (PubMed)
  • Download PDF
  • Download XML
  • View XML
Review Open Access

PANoptosis: A novel therapeutic target in kidney disease (Review)

  • Authors:
    • Lianyan Jiang
    • Liangbin Zhao
    • Yu Liu
    • Zhongmei Fu
    • Mengzhu Wu
    • Zengyi Mou
    • Mingquan Li
  • View Affiliations / Copyright

    Affiliations: School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610032, P.R. China, Department of Nephrology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, P.R. China, Department of Nephrology, The Chinese University of Hong Kong, Shenzhen Medical Centre, Shenzhen, Guangdong 518100, P.R. China
    Copyright: © Jiang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
  • Article Number: 207
    |
    Published online on: September 26, 2025
       https://doi.org/10.3892/ijmm.2025.5648
  • Expand metrics +
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Metrics: Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )
Cited By (CrossRef): 0 citations Loading Articles...

This article is mentioned in:



Abstract

The global prevalence of kidney diseases continues to rise annually, posing a serious public health challenge, and notably contributing to morbidity and mortality rates worldwide. In affected individuals, kidney function progressively declines, ultimately resulting in renal failure. Therefore, elucidating the pathophysiological mechanisms underlying kidney diseases and identifying novel therapeutic targets is of paramount importance. Among these, the regulation of cell death pathways has emerged as a pivotal factor in disease progression. PANoptosis, a recently characterized form of programmed cell death, is activated by diverse stimuli, and involves the integrated activation of pyroptosis, apoptosis and necroptosis, coordinated through the PANoptosome complex. Increasing evidence has suggested that PANoptosis serves an important role in the development of kidney diseases, and further investigation into its regulatory mechanisms may provide valuable insights into disease modulation. The present review summarizes current knowledge regarding the mechanisms of PANoptosis and its involvement in various kidney diseases, offering perspectives for future diagnostic and therapeutic strategies.

Introduction

Programmed cell death (PCD) is a tightly regulated biological process influenced by various external stimuli and intracellular conditions. PCD encompasses several distinct forms of cell death, including apoptosis, pyroptosis, necroptosis, ferroptosis and autophagy, each mediated through specific genetic and molecular mechanisms (1). These forms of cell death are essential for embryonic development, the maintenance of tissue homeostasis and the clearance of intracellular pathogens, and are regarded as fundamental processes for preserving physiological stability (1,2). PCD pathways can be categorized into lytic and non-lytic forms of cell death. Lytic cell death, such as pyroptosis and necroptosis, involves membrane potential loss and cellular swelling, resulted in disrupted cell integrity and ultimately leading to cell rupture, releasing a large amount of inflammatory mediators (2). Comparatively, non-lytic forms of PCD, such as apoptosis, involve the coordinated breakdown of dying cells into smaller fragments for the isolation of cellular contents, which limits the release of cytokines and damage-associated molecular patterns (DAMPs), typically in a silent manner without triggering a pronounced immune response (1).

PANoptosis is a recently identified form of PCD that is initiated by innate immune signaling, and integrates key features of pyroptosis, apoptosis and necroptosis. Notably, its biological effects cannot be fully explained by any of these pathways alone. The concept of PANoptosis was first introduced by Malireddi et al in 2019 (3) (Fig. 1). Mechanistically, PANoptosis is mediated through the assembly of a multiprotein complex known as the PANoptosome, which incorporates essential components from the pyroptotic, apoptotic and necroptotic pathways. This complex enables cross-talk and coordinated activation among the three pathways, resulting in a unified mode of cell death (4,5). Due to the regulatory role of the PANoptosome, inhibition of a single form of cell death is insufficient to prevent PANoptosis. Instead, effective blockade requires targeting components within the PANoptosome complex.

Timeline of the discovery of
apoptosis, necroptosis, pyroptosis and PANoptosis. In 2019,
PANoptosis was first named, and four types of PANoptosomes have
since been characterized. Created with MedPeer (medpeer.cn).
NLRP12, Nod-like receptor pyrin domain-containing protein 12;
RIPK1, receptor-interacting protein kinase 1; ZBP1, Z-DNA binding
protein 1.

Figure 1

Timeline of the discovery of apoptosis, necroptosis, pyroptosis and PANoptosis. In 2019, PANoptosis was first named, and four types of PANoptosomes have since been characterized. Created with MedPeer (medpeer.cn). NLRP12, Nod-like receptor pyrin domain-containing protein 12; RIPK1, receptor-interacting protein kinase 1; ZBP1, Z-DNA binding protein 1.

Emerging evidence has revealed that PANoptosis contributes to the pathogenesis of several kidney diseases, including both acute and chronic forms (6-12). Although the role of PANoptosis has been extensively studied in cancer, infections and inflammatory conditions, its function in renal pathology remains insufficiently characterized. The present review summarizes the molecular mechanisms underlying PANoptosis activation and explores its potential involvement in acute kidney injury (AKI), chronic kidney disease (CKD), diabetic kidney disease (DKD), renal cell carcinoma (RCC) and drug-induced kidney injury, aiming to provide a theoretical foundation for PANoptosis-targeted therapeutic strategies in kidney disease.

Literature search, and cross-regulation of apoptosis, pyroptosis and necroptosis

A comprehensive search for studies published up to May 2025 was performed on platforms including PubMed (https://pubmed.ncbi.nlm.nih.gov/), Google Scholar (https://scholar.google.com/), Web of Science (https://webofscience.com/WOS), ScienceDirect (https://www.sciencedirect.com/) and CNKI (https://www.cnki.net/). The search adopted a combination of subject terms and free terms, and was appropriately adjusted according to the rules of the different databases. The key terms included 'Kidney disease', 'Kidney failure', 'Renal insufficiency', 'Renal failure', 'Nephropathy', 'Renal disease' and 'PANoptosome' or 'PANoptosis'. Notably, low-quality articles and papers with incomplete information were excluded. The main inclusion criteria for the literature were original research or comprehensive reports. The research content clearly involved the mechanism or therapeutic significance of PANoptosis in kidney diseases, and the research subjects included humans and mammals. The screening was independently conducted by two researchers, and any disputes arising during the process were resolved through mutual discussion or arbitration by a third-party senior researcher.

Apoptosis

Apoptosis, pyroptosis and necroptosis represent the three primary forms of PCD, each characterized by distinct initiators, signaling mediators and execution mechanisms. Among them, apoptosis is the most extensively studied and was first described by Kerr et al in 1972 (13) (Fig. 1). Apoptosis depends on the involvement of caspases. Morphologically, apoptosis is defined by characteristic features including cell shrinkage, cytoplasmic condensation, membrane blebbing and nuclear chromatin condensation (13,14). Apoptosis occurs through two main pathways: The intrinsic (mitochondrial) pathway and the extrinsic (death receptor-mediated) pathway (Fig. 2A). The intrinsic pathway is activated in response to intracellular stressors such as DNA damage, oxidative stress or toxic insults. In this context, key stress-sensing proteins, such as the tumor suppressor protein 53 (P53), serve a central regulatory role by transcriptionally activating pro-apoptotic members of the Bcl-2 family, such as BAK and BAX, while concurrently suppressing anti-apoptotic proteins, such as Bcl-2 and Bcl-XL (15-17). Activated BAK and BAX oligomerize on the mitochondrial outer membrane, resulting in mitochondrial outer membrane permeabilization and the release of cytochrome c (Cyt c) into the cytosol (18). Cyt c subsequently binds to apoptotic protease-activating factor 1, promoting the formation of the apoptosome complex, which leads to the activation of caspase-9 (19,20). Caspase-9 then cleaves and activates downstream effector caspases, including caspase-3 and caspase-7, ultimately executing the apoptotic process (21). The extrinsic pathway is initiated by the binding of death ligands to their corresponding receptors. Fas and tumor necrosis factor receptor (TNFR) recruit adaptor proteins, such as TNFR-associated death domain protein (TRADD) and Fas-associated death domain protein (FADD), forming the death-inducing signaling complex, which activates caspase-8 and caspase-10 (22-24). Active caspase-8/10 cleaves and activates the effector caspases, including caspase-3, caspase-7 and caspase-6, leading to apoptosis (25,26). Caspase-8 also cleaves BID, which activates the intrinsic pathway and further promotes apoptosis (27).

Mechanisms of apoptosis, necroptosis
and pyroptosis. (A) Apoptosis depends on the involvement and
regulation of caspases, and is divided into intrinsic (dependent on
caspase-9) and extrinsic (dependent on caspase-8/10) pathways,
which respectively cleave downstream factors dependent on
caspase-3/6/7 to perform apoptosis. (B) RIPK1 recruits and
phosphorylates RIPK3 in necroptosis, further activating MLKL. MLKL
has pore-forming activities, leading to necroptosis. (C) Pyroptosis
is divided into classical (dependent on caspase-1) and
non-classical (dependent on caspase-4/5/11) pathways, which rely on
the cleavage of the executive protein GSDMD, forming N-terminal
GSDMD fragments that form pores in the cell membrane, inducing the
release of inflammatory exudate and driving pyroptosis. The three
modes of cell death are not isolated, but interact with each other,
for example, caspase-8 is involved in regulating all three cell
death modes simultaneously. Created with MedPeer (medpeer.cn).
APAF-1, apoptotic protease-activating factor 1; ASC,
apoptosis-related speck-like protein; Cyt c, cytochrome c;
DAMPs, damage-associated molecular patterns; DISC, death-inducing
signaling complex; FADD, Fas-associated death domain protein;
GSDMD, gasdermin D; LPS, lipopolysaccharide; MLKL, mixed-lineage
kinase domain-like protein; MOMP, mitochondrial outer membrane
permeabilization; NLRP3, Nod-like receptor pyrin domain-containing
protein 3; PAMPs, pathogen-associated molecular patterns; RIPK,
receptor-interacting protein kinase; ROS, reactive oxygen species;
TNF, tumor necrosis factor; TNFR, TNF receptor; TRADD,
TNFR-associated death domain protein; TRAIL, TNF-related
apoptosis-inducing ligand; TRAILR, TRAIL receptor.

Figure 2

Mechanisms of apoptosis, necroptosis and pyroptosis. (A) Apoptosis depends on the involvement and regulation of caspases, and is divided into intrinsic (dependent on caspase-9) and extrinsic (dependent on caspase-8/10) pathways, which respectively cleave downstream factors dependent on caspase-3/6/7 to perform apoptosis. (B) RIPK1 recruits and phosphorylates RIPK3 in necroptosis, further activating MLKL. MLKL has pore-forming activities, leading to necroptosis. (C) Pyroptosis is divided into classical (dependent on caspase-1) and non-classical (dependent on caspase-4/5/11) pathways, which rely on the cleavage of the executive protein GSDMD, forming N-terminal GSDMD fragments that form pores in the cell membrane, inducing the release of inflammatory exudate and driving pyroptosis. The three modes of cell death are not isolated, but interact with each other, for example, caspase-8 is involved in regulating all three cell death modes simultaneously. Created with MedPeer (medpeer.cn). APAF-1, apoptotic protease-activating factor 1; ASC, apoptosis-related speck-like protein; Cyt c, cytochrome c; DAMPs, damage-associated molecular patterns; DISC, death-inducing signaling complex; FADD, Fas-associated death domain protein; GSDMD, gasdermin D; LPS, lipopolysaccharide; MLKL, mixed-lineage kinase domain-like protein; MOMP, mitochondrial outer membrane permeabilization; NLRP3, Nod-like receptor pyrin domain-containing protein 3; PAMPs, pathogen-associated molecular patterns; RIPK, receptor-interacting protein kinase; ROS, reactive oxygen species; TNF, tumor necrosis factor; TNFR, TNF receptor; TRADD, TNFR-associated death domain protein; TRAIL, TNF-related apoptosis-inducing ligand; TRAILR, TRAIL receptor.

Necroptosis

Necroptosis was first reported by Degterev et al (28) in 2005 as a distinct form of PCD (Fig. 1). It is morphologically characterized by organelle swelling, plasma membrane rupture and leakage of cellular contents (29,30) and is regulated through the formation of the necrosome, a signaling complex composed of receptor-interacting protein kinase 1/3 (RIPK1/3) and mixed-lineage kinase domain-like protein (MLKL) (31,32) (Fig. 2B). Upon stimulation or injury, death receptors such as TNFR1, Fas, TNF and TNF-related apoptosis-inducing ligand (TRAIL) receptor bind to their respective ligands, initiating downstream signaling cascades. These interactions recruit kinases and adaptor proteins that assemble the necrosome complex (33-36). Within the necrosome, RIPK1 activates RIPK3 through phosphorylation (37), and this activation frequently leads to NF-κB-mediated pro-inflammatory and pro-survival signaling (35,38,39). When caspase-8 is active, it forms a complex with RIPK1 and FADD to initiate apoptosis. However, when caspase-8 is inhibited, RIPK1 engages and phosphorylates RIPK3 via the RIP homotypic interaction motif (RHIM) domain. Activated RIPK3 subsequently phosphorylates MLKL, which oligomerizes and translocates to the plasma membrane. MLKL is the terminal effector of necroptosis (40-43). Oligomerized MLKL disrupts membrane integrity by forming pores, leading to ion imbalance and water influx, which cause cell swelling and rupture (44,45). This facilitates the release of DAMPs into the extracellular space, activating the innate immune response, and amplifying inflammation and tissue injury (46).

Pyroptosis

Pyroptosis was first observed in 1992 when macrophages infected with Shigella flexneri exhibited cell death characterized by features initially mistaken for apoptosis (47). Subsequent studies revealed that this process was distinct from apoptosis and relied on caspase-1 activation rather than the classical apoptotic molecule caspase-3 (48). In 2001, Cookson and Brennan (49) introduced the term 'pyroptosis' to describe this caspase-1-dependent form of PCD (Fig. 1).

Pyroptosis is an inflammation-associated form of PCD that occurs through the classical or non-classical pathways. While this process shares some morphological features with apoptosis, such as nuclear condensation and DNA fragmentation, its hallmark characteristics include pore formation in the plasma membrane, cell swelling, membrane rupture and the release of pro-inflammatory intracellular contents; these events trigger a robust inflammatory response (49-51). Upon exposure to toxic stimuli, intracellular and extracellular danger signals induce the formation of inflammasome complexes in the cytoplasm through the classical caspase-1-dependent pathway or the non-classical caspase-4/5/11-dependent pathway; these inflammasome complexes are composed of Nod-like receptor family pyrin domain containing (NLRP)3/NLRP1, apoptosis-related speck-like protein (ASC) and pro-caspase-1 (Fig. 2C). Inflammasomes promote the production of caspase-1, and the maturation and secretion of IL-1β and IL-18, and induce cleavage of the pyroptosis executioner protein gasdermin D (GSDMD). The N-terminal fragment of GSDMD forms pores in the membrane, initiating pyroptosis (51-54).

Interaction and cross-regulation among apoptosis, pyroptosis, and necroptosis

For a long time, different cell death modalities were examined in isolation due to their regulation by distinct molecular mechanisms. However, the identification of PANoptosis has revealed that these pathways are interconnected at multiple levels and can be activated simultaneously or sequentially within the same cell. Rather than acting independently, these forms of cell death can switch from one to another under specific conditions, highlighting their close association and mutual regulation (3,4).

Among the key mediators linking these pathways, caspases have a central role. Caspase-8, classically recognized as the initiator enzyme of the extrinsic apoptotic pathway, promotes the cleavage of BID to tBID, which subsequently activates BAX and BAK, and initiates apoptotic signaling. However, increasing evidence (22,55-60) has indicated that caspase-8 also participates in necroptosis and pyroptosis (Fig. 2B and C); it can trigger necroptosis mediated by RIPK3 and MLKL, and also contributes to pyroptotic regulation (55-58). Caspase-8 forms an oligomeric complex with RIPK1 and FADD, which facilitates the activation of downstream effector caspases-3/7 and promotes apoptosis (59,60). Additionally, caspase-8 can directly cleave RIPK1 and RIPK3, thereby suppressing necroptosis. When caspase-8 activity is inhibited, this suppression is lifted, allowing RIPK3 activation and promoting necroptotic cell death (61,62). RIPK1 also contributes to FADD/caspase-8-dependent apoptosis. Upon TNFR1 engagement, the TNFR1 signaling complex recruits TRADD, which subsequently recruits FADD. FADD then binds pro-caspase-8, facilitates its dimerization and activation, and initiates apoptosis (63). Dillon et al (64) demonstrated through both in vitro and in vivo studies that RIPK1 can regulate FADD/caspase-8-mediated apoptosis as well as RIPK3/MLKL-induced necroptosis, thereby preventing perinatal mortality in mice (Fig. 2A and B). Caspase-8 also has a regulatory role in pyroptosis by facilitating activation of the NLRP3 inflammasome, promoting ASC complex formation and caspase-1 activation, which in turn induces IL-1β secretion and pyroptosis (55,62). In the context of Yersinia infection, inhibition or deletion of transforming growth factor-β-activated kinase 1 (TAK1) has been shown to enhance caspase-8-mediated GSDMD cleavage, thereby favoring pyroptosis while reducing apoptosis (65) (Fig. 2A and C). Furthermore, caspase-8 can act on other GSDM family members, such as GSDMC, which is specifically cleaved under TNFα stimulation to generate the N-terminal domain of GSDMC, leading to pore formation in the plasma membrane and the induction of pyroptosis (66,67). During influenza virus infection, caspase-8 activates caspase-3, which cleaves GSDMD into an inactive form, thereby reducing IL-1β and IL-18 release, and suppressing pyroptosis (68). These findings collectively indicate that caspase-8 is a pivotal regulator linking apoptosis, necroptosis and pyroptosis.

Caspase-3, a key executioner of apoptosis, also contributes to pyroptosis. Upon activation, it cleaves GSDME, generating the GSDME-N fragment that induces pyroptotic membrane disruption (69,70). Similarly, caspase-1, the terminal effector of pyroptosis, has been shown to mediate apoptosis in the absence of GSDMD by activating the BID/caspase-9/caspase-3 axis (71,72).

In addition to caspases, multiple signaling complexes and inflammasomes contribute to the cross-talk among these cell death pathways. NLRP3, the most extensively studied inflammasome, serves a critical role in pyroptosis by promoting inflammatory responses. Potassium efflux, a known activator of NLRP3, can result from MLKL-mediated membrane disruption during necroptosis, which subsequently leads to caspase-1 activation and IL-1β maturation (73-75) (Fig. 2B and C). Moreover, upregulation of RIPK3 has been shown to trigger the RIPK3/MLKL/NLRP3/caspase-1 signaling cascade, further supporting the interconnected nature of these pathways (76).

Collectively, these findings indicate that apoptosis, pyroptosis and necroptosis constitute an interconnected network rather than three independent cell death pathways, with key regulators, such as caspase-8, exerting multiple functions, including promoting apoptosis through activation of caspase-3/7, suppressing necroptosis by cleaving RIPK1 and RIPK3, and participating in NLRP3 inflammasome-dependent pyroptosis. Similarly, RIPK1 is involved in apoptosis and pyroptosis. These pathways are not isolated but rather interdependent, with key molecules functioning as receptors for initiating and recognizing death signals, as adapters for recruiting downstream mediators, and as effectors that execute the final cell death program.

Mechanism and key molecules involved in PANoptosis

The concept of PANoptosis emerged from studies investigating the molecular overlap among pyroptosis, apoptosis and necroptosis. Increasing evidence has suggested that these three forms of PCD can occur simultaneously and are interconnected through a complex molecular network, rather than acting as isolated pathways (77-80), leading to the recognition of PANoptosis as a distinct, coordinated form of cell death. PANoptosis is a newly characterized PCD pathway regulated by innate immune responses and mediated through the assembly of a multiprotein complex known as the PANoptosome. It is initiated by innate immune sensors and executed via caspases and RIPKs. The PANoptosome integrates components from the pyroptotic, apoptotic and necroptotic machinery, enabling their coordinated activation. Regulation of the PANoptosome allows simultaneous control of all three death pathways, while also providing a molecular scaffold that facilitates interaction among key signaling molecules, thereby driving PANoptosis (81,82).

Activation of PANoptosis involves upstream receptors and signaling cascades that lead to the formation of the PANoptosome. These complexes serve as platforms for cross-talk among the three PCD pathways and are essential for their integration (83). The PANoptosome typically comprises three categories of components (5,8,11,80,83): i) Pattern recognition receptors or sensors that detect pathogen-associated molecular patterns (PAMPs) and DAMPs, including Z-DNA binding protein 1 (ZBP1), NLRP12, AIM2, RIPK1, pyrin and NLRP3; ii) adaptor proteins such as ASC and FADD; and iii) catalytic effectors, including RIPK1, RIPK3, caspase-1 and caspase-8. The composition of the PANoptosome is context-dependent and varies according to the nature of the stimulus or pathological condition. In general, the sensors recognize PAMPs or DAMP,s and initiate PANoptosome assembly through homotypic or heterotypic interactions among adaptor domains. This assembly provides a structural platform for the convergence of apoptotic, pyroptotic and necroptotic effectors. The resulting complex enables the coordinated activation of caspases and RIPKs, thereby inducing pyroptosis, apoptosis and necroptosis in a synchronized manner (80,83).

The PANoptosome complexes characterized to date include the ZBP1-PANoptosome (84-86), AIM2-PANoptosome (82), RIPK1-PANoptosome (87) and NLRP12-PANoptosome (88) (Figs. 1 and 3). Among them, ZBP1 is a critical innate immune sensor involved in host defense that serves as a key initiator of PANoptosis. Structurally, ZBP1 contains two N-terminal Zα domains (Zα1 and Zα2), two RHIMs (RHIM1 and RHIM2) and a C-terminal domain. The expression of ZBP1 is typically induced by interferons (IFNs), and ZBP1 is also activated in response to pathogens or cellular stress (3,86,89). In 2016, Kuriakose et al (89) identified ZBP1 as a shared regulator of apoptosis, pyroptosis, and necroptosis. Later, in 2019, the same group demonstrated that ZBP-1 and TAK1 can initiate the assembly of PANoptosome complexes (3). In 2020, the ZBP1-PANoptosome was further characterized (84,85). This complex orchestrates the induction of cytokine release along with the activation of apoptosis, pyroptosis and necroptosis, contributing to pathogen clearance and the maintenance of immune homeostasis (3). The ZBP1-PANoptosome is composed of RIPK3, caspase-8, caspase-6, caspase-1, RIPK1, ZBP1, ASC and NLRP3 (84-86) (Fig. 3A). During influenza A virus (IAV) infection, the RHIM domain of ZBP1 engages in homotypic interactions with the RHIM domains of RIPK1 and RIPK3, facilitating the recruitment of caspase-6 and/or caspase-8, and activating ASC, NLRP3 and caspase-1 to assemble the PANoptosome (85). This complex promotes necroptosis via the RIPK3/MLKL axis, induces pyroptosis through NLRP3 inflammasome-mediated GSDMD cleavage, and activates apoptosis through caspase-8 and downstream effectors, such as caspase-3 and caspase-7 (90-93). Additionally, caspase-6 enhances the ZBP1-RIPK3 interaction during IAV infection and contributes to NLRP3 inflammasome activation, further facilitating PANoptosome assembly (78,94). The Zα2 domain of ZBP1 serves a vital role in sensing Z-RNAs within infected cells and mediating inflammasome activation during IAV infection, thereby promoting PANoptosis (85,93). Similar mechanisms have been observed in infections caused by coronaviruses and mouse hepatitis virus, where ZBP1 activation contributes to cytokine storm and PANoptosis (86). Deletion of ZBP1 in β-coronavirus-infected mice has been shown to reduce lung injury, and to markedly suppresses cytokine storm and PANoptosis (86,95). Beyond viral infections, ZBP1 is also critical in PANoptosis induced by fungal and bacterial pathogens. It acts as an upstream sensor during fungal infections and initiates PANoptosis (84). In Francisella novicida infection, ZBP1 contributes to the assembly of the AIM2-PANoptosome, interacting with AIM2, pyrin, ASC and other components (82). However, in Yersinia-induced PANoptosis, ZBP1 is not essential, as this process is instead dependent on RIPK1, highlighting the involvement of distinct innate immune sensors under different pathological conditions (87).

Molecular compositions and mechanisms
of the PANoptosome complexes. (A) ZBP1-PANoptosome consists of
RIPK3, caspase-8, caspase-6, caspase-1, RIPK1, ZBP1, ASC and NLRP3.
(B) AIM2-PANoptosome is assembled by combining molecules such as
AIM2, ZBP1, pyrin, ASC, caspase-8, caspase-1, RIPK3, RIPK1, NLRP3
and FADD. (C) RIPK1-PANoptosome contains molecules such as
caspase-8, caspase-1, FADD, NLRP3, ASC, RIPK1 and RIPK3. (D)
NLRP12-PANoptosome consists of NLRP12, caspase-1, caspase-8, RIPK3,
NLRP3 and ASC. The four PANoptosomes all contain key factors for
three cell death modes, caspase-3/7, MLKL and GSDMD, which induce
apoptosis, necroptosis and pyroptosis, respectively. Created with
MedPeer (medpeer. cn). AKI, acute kidney injury; ASC,
apoptosis-related speck-like protein; CKD, chronic kidney disease;
DAMPs, damage-associated molecular patterns; FADD, Fas-associated
death domain protein; GSDMD, gasdermin D; HSV1, herpes simplex
virus 1; IAV, influenza A virus; MLKL, mixed-lineage kinase
domain-like protein; NLRP, Nod-like receptor family pyrin domain
containing; PAMPs, pathogen-associated molecular patterns; RCC,
renal cell carcinoma; RIPK, receptor-interacting protein kinase;
TAK1, transforming growth factor-β-activated kinase 1; TLR,
Toll-like receptor; ZBP1, Z-DNA binding protein 1.

Figure 3

Molecular compositions and mechanisms of the PANoptosome complexes. (A) ZBP1-PANoptosome consists of RIPK3, caspase-8, caspase-6, caspase-1, RIPK1, ZBP1, ASC and NLRP3. (B) AIM2-PANoptosome is assembled by combining molecules such as AIM2, ZBP1, pyrin, ASC, caspase-8, caspase-1, RIPK3, RIPK1, NLRP3 and FADD. (C) RIPK1-PANoptosome contains molecules such as caspase-8, caspase-1, FADD, NLRP3, ASC, RIPK1 and RIPK3. (D) NLRP12-PANoptosome consists of NLRP12, caspase-1, caspase-8, RIPK3, NLRP3 and ASC. The four PANoptosomes all contain key factors for three cell death modes, caspase-3/7, MLKL and GSDMD, which induce apoptosis, necroptosis and pyroptosis, respectively. Created with MedPeer (medpeer. cn). AKI, acute kidney injury; ASC, apoptosis-related speck-like protein; CKD, chronic kidney disease; DAMPs, damage-associated molecular patterns; FADD, Fas-associated death domain protein; GSDMD, gasdermin D; HSV1, herpes simplex virus 1; IAV, influenza A virus; MLKL, mixed-lineage kinase domain-like protein; NLRP, Nod-like receptor family pyrin domain containing; PAMPs, pathogen-associated molecular patterns; RCC, renal cell carcinoma; RIPK, receptor-interacting protein kinase; TAK1, transforming growth factor-β-activated kinase 1; TLR, Toll-like receptor; ZBP1, Z-DNA binding protein 1.

AIM2 is an inflammasome sensor belonging to the PYHIN protein family that recognizes cytoplasmic double-stranded DNA (dsDNA). Structurally, AIM2 contains an N-terminal pyrin domain and a C-terminal HIN200 domain, and is primarily localized in the cytoplasm (96-98). Upon detection of pathogen-derived dsDNA, AIM2 initiates the formation of the AIM2-PANoptosome complex. This assembly activates the inflammasome pathway, leading to the secretion of IL-1β and IL-18, and inducing pyroptosis (99,100). In bone marrow-derived macrophage models infected with herpes simplex virus 1 and Francisella novicida, AIM2 has been shown to regulate other innate immune sensors, including pyrin and ZBP1. Together with ASC, caspase-1, caspase-8, RIPK3, RIPK1, NLRP3 and FADD, these components assemble to form the AIM2-PANoptosome, driving PANoptosis (82) (Fig. 3B). During infection, AIM2 deficiency markedly reduces inflammation and cell death, whereas the absence of pyrin or ZBP1 produces a partial reduction in these responses. These observations suggest that AIM2 functions as an upstream regulator controlling the assembly and activation of the AIM2-PANoptosome (82).

RIPK1 is another RHIM-containing protein that serves a central role in inflammation, cell death and the regulation of PANoptosis. It can activate necroptosis by promoting the phosphorylation of RIPK3, which interacts with MLKL to induce cell death through the RIPK3/MLKL axis (40,89,101). Dillon et al (64) reported that RIPK1 also contributes to FADD/caspase-8-mediated apoptosis and RIPK3-dependent necroptosis. TAK1 serves as a key upstream regulator of RIPK1, controlling the formation and activation of PANoptosomes (3,102,103). Under normal physiological conditions, TAK1 inhibits TNF-mediated autocrine signaling by suppressing TNF production and RIPK1 phosphorylation, thereby preventing PANoptosis activation (28,79). In Yersinia-infected cells, TAK1 inhibition or knockout promotes the assembly of the RIPK1-PANoptosome, which includes RIPK1, caspase-8, caspase-1, FADD, NLRP3, ASC and RIPK3 (Fig. 3C). This complex activates FADD-Caspase-8-mediated apoptosis, RIPK3-MLKL-dependent necroptosis, and NLRP3 inflammasome-mediated pyroptosis (79,87). Moreover, in the absence of TAK1, microbial signals can activate PANoptosis through the RIPK3/MLKL axis independent of RIPK1 kinase activity (3). Using models specifically lacking TAK1, studies have demonstrated that inhibition of RIPK1 kinase activity in TAK1-deficient bone marrow-derived macrophages can suppress PANoptosis and inflammation, infection and myeloid proliferation (77,79,104,105).

NLRP12 is a member of the NLR family and contains an N-terminal pyrin domain, a central nucleotide-binding domain and a C-terminal leucine-rich repeat region. It functions as an innate immune sensor that modulates inflammatory signaling in response to infection and cellular stress (106). It has been demonstrated that heme, in conjunction with PAMPs, activates the Toll-like receptor 2/4 signaling axis, which induces the expression of NLRP12 (88). This induction promotes the assembly of a PANoptosome complex composed of NLRP12, NLRP3, ASC, caspase-1, caspase-8 and RIPK3, collectively referred to as the NLRP12-PANoptosome (Fig. 3D). Activation of this complex leads to PANoptosis, characterized by caspase-1, caspase-8 and caspase-3 activation, MLKL phosphorylation, and cleavage of GSDMD or GSDME. A functional study further revealed that NLRP12 deficiency confers protection against heme-induced AKI and mortality in mice, underscoring its critical role in disease pathogenesis (88). This finding suggests that NLRP12 and associated PANoptotic components may represent promising therapeutic targets for hemolytic and inflammation-related disorders (88).

The role of PANoptosis in kidney diseases

AKI

AKI is a clinical syndrome characterized by a sudden decline in renal function, and is associated with a high incidence and poor clinical outcomes (107,108). The incidence of AKI affects ~20% of hospitalized patients and exceeds 50% among those in intensive care units, making it a major contributor to in-hospital mortality (109). Emerging evidence (8,110-112) has indicated that PANoptosis serves an important role in various AKI models, including those induced by sepsis, ischemia/reperfusion (I/R) injury and exposure to nephrotoxic agents, such as chemicals and certain traditional Chinese medicines.

Cell death and inflammation are central events in the pathogenesis of AKI, with I/R injury being a primary cause. Previous studies have demonstrated that apoptosis, pyroptosis and necroptosis coexist in models of middle cerebral artery occlusion, oxygen-glucose deprivation/recovery and retinal I/R injury (113,114). Another study, by collecting data from animal and cell experiments, reported that PANoptosis is observed in ischemic brain injury (115). These findings suggest that PANoptosis may represent a promising therapeutic target for diseases associated with I/R injury. In I/R-induced AKI, renal tubular epithelial cells experience endoplasmic reticulum stress, oxidative stress and inflammation due to hypoxia, followed by reperfusion, leading to acute tubular cell loss. Numerous animal studies and analyses of kidney biopsies from transplant recipients have demonstrated that I/R induces apoptosis in tubular epithelial cells (116-118). Although apoptosis was initially regarded as the principal mechanism of AKI, further studies have shown that multiple cell death pathways are involved, and inhibiting apoptosis alone is insufficient to prevent renal injury (119). In I/R-induced AKI models, NLRP3 activates caspase-1, leading to GSDMD cleavage and the release of the pro-inflammatory cytokines IL-1β and IL-18, thereby inducing pyroptosis in renal tubular epithelial cells (120-122). Pyroptosis has also been observed in AKI models caused by cisplatin and sepsis (123). Linkermann et al (124,125) reported increased expression of RIPK1 and RIPK3 in I/R-induced AKI, and demonstrated that treatment with the necroptosis inhibitor necrostatin-1 (Nec-1) markedly improved renal function. Similarly, the RIPK1 inhibitor Compound-71 has been shown to alleviate cisplatin-induced AKI by suppressing necroptosis (126). In calcium oxalate-induced AKI, ZBP1 activation of the RIPK3/MLKL axis mediates necroptosis, whereas ZBP1 knockout can reduce necroptotic activity and tubular injury (127). These findings underscore the importance of simultaneously targeting apoptosis, pyroptosis and necroptosis to protect against AKI-associated kidney damage.

While previous studies have often focused on individual PCD pathways, the discovery of PANoptosis has shifted attention to the integrated role of multiple death mechanisms in AKI. In a mouse model of sepsis-induced AKI established by cecal ligation and puncture, increased AIM2 expression in kidney tissue has been shown to be associated with concurrent activation of apoptosis, pyroptosis, necroptosis and inflammasome signaling, supporting the presence of PANoptosis (110). In vitro, PANoptosis has been observed in lipopolysaccharide (LPS)-treated human proximal tubular epithelial HK-2 cells, and AIM2 silencing may reverse this effect, suggesting that AIM2 mediates sepsis-induced renal injury through regulation of PANoptosis (110). NLRP3, a key component of the four types of PANoptosome, has also been shown to be involved in I/R-induced AKI. The clinical agent 3,4-methylenedioxy-β-nit rostyrene has been report to reduce apoptosis, necroptosis and pyroptosis in a renal I/R injury model by targeting NLRP3, thereby alleviating tissue damage (8). Furthermore, an aqueous extract of Achyranthes aspera can ameliorate cisplatin-induced AKI by inhibiting DNA damage, oxidative stress, inflammation and PANoptosis (111). In sepsis-induced AKI mouse models, elevated levels of PANoptosis-related molecules, including RIPK1, MLKL, caspase-3/7 and FADD, have been observed in renal tissue, along with marked upregulation of ZBP1, particularly in the renal interstitium, highlighting the essential role of PANoptosis in the pathogenesis of sepsis-induced AKI (112) (Table I).

Table I

PANoptosis in various kidney diseases.

Table I

PANoptosis in various kidney diseases.

First author, yearDiseaseTriggerExperimental methodDamaged cells/tissues Target/participation factors(Refs.)
Wei, 2024AKICLPIn vivo/in vitroRTECsAIM2, caspase-1, caspase-8, RIPK1, MLKL, ZBP1(110)
Ruilian, 2023AKILPSIn vitroRenal cellsFADD, caspase-3, caspase-7, caspase-8, RIPK1, MLKL, ZBP1(112)
Lin, 2024AKICisplatinIn vivoRenal cellsNLRP3, caspase-1, caspase-3, NRF2, Bcl-2(111)
Uysal, 2022AKIRIRIn vitroRTECsGSDMD, caspase-3, MLKL, NLRP3(8)
Mall, 2022KIRCNABioinformatics methodsNAHigh expression of PANoptosis-related genes, such as AIM2, caspase-3, caspase-4 and TNFRSF10, is significantly associated with poor prognosis in KIRC(132)
Mall, 2023KIRCNABioinformatics methodsNAGene mutation patterns, activation of specific immune cells and oncogenic pathways are significantly different between high and low PANoptosis cluster groups(7)
Jiang, 2024KIRCNABioinformatics methodsNAPANoptosis has a regulatory effect on the tumor immune microenvironment in KIRC and serves an important role in the development of KIRC(9)
Wang, 2023ccRCC786-O and Caki-1 cellsBioinformatics methods/in vitroccRCC cellsThe signal constructed based on PANoptosis-related microRNAs has clinical significance for predicting the progression of ccRCC(133)
Liu, 2023ccRCC786-O and Caki-1 cellsCombining bioinformatics and in vitro methodsccRCC cellsPANoptosis-related long non-coding RNAs are associated with the progression, prognosis, immune regulation and therapeutic efficacy of ccRCC(134)
Xu, 2024KIRC, KIRP and KICH769-P and 786-O cellsCombining bioinformatics and in vitro methodsKIRC, KIRP and KICH cellsThe new indicator PANII constructed based on PANoptosis-related genes (BAX, caspase-1, caspase-8 and PYCARD) can reflect the immune regulatory status and predict immune therapy response(135)
Zhang, 2024CKDAdenineCombining bioinformatics and in vivo methodsRTECsFOS and PTGS2 genes may be closely related to the pathological mechanism of PANoptotic CKD(150)
Yang, 2025CKDSevere burnsIn vivo/in vitroMacrophages and podocytesCaspase-1, caspase-3, IL-1β, GSDMD(155)
Lv, 2025DKDSTZIn vivo/in vitroPodocytesRIPK1, tMLKL, Bcl-2, BAX, caspase-3, caspase-1(156)
Yi, 2024Kidney damage caused by drugsATRIn vivoRenal cellsLYC inhibits ATR exposure-induced renal PANoptosis and inflammation (AIM2, caspase 1, GSDMD, MLKL, pyrin, ZBP1)(10)
Zhang, 2023Renal injury caused by drugsTriptolideIn vivo/in vitroMacrophagesASC, RIPK3, caspase-8, GSDMD, MLKL, caspase-3, caspase-1(157)
Xu, 2024Renal injury caused by drugsAristolochic acidIn vivo/in vitroRTECsRIPK3, GSDMD, MLKL, caspase-3, caspase-8, ASC(158)
Li, 2024Renal injury caused by drugsCBL0137 + LPSIn vivo/in vitroMacrophagesZBP1, ASC, NLRP3, GSDMD, MLKL, caspase-1(11)
Xie, 2024Renal injury caused by drugs TrichloroethyleneIn vivo/in vitroEndothelial cellsRIPK1, GSDMD, MLKL, caspase-1, caspase-3, caspase-8, ASC, RIPK3, NLRP3(6)
Yuan, 2022Kidney damage caused by other factorsHeatstrokeIn vivo/in vitroBMDMs, peritoneal macrophages and L929 mouse fibroblastsZBP1, GSDMD, MLKL, caspase-3, caspase-8, RIPK1, RIPK3(159)

[i] AKI, acute kidney injury; ATR, atrazine; BMDMs, bone marrow-derived macrophages; CKD, chronic kidney disease; CLP, cecal ligation and puncture; DKD, diabetic kidney disease; FADD, Fas-associated death domain protein; GSDMD, gasdermin D; KIRC/ccRCC, clear cell RCC; KIRC, kidney renal clear cell carcinoma; KIRP, papillary RCC; KICH, chromophobe RCC; LPS, lipopolysaccharide; MLKL, mixed-lineage kinase domain-like protein; NLRP, Nod-like receptor pyrin domain-containing protein; PANII, PANoptosis-Immunity Index; RCC, renal cell carcinoma; RIPK, receptor-interacting protein kinase; RTECs, renal tubular epithelial cells; STZ, streptozotocin; ZBP1, Z-DNA binding protein 1; ATR, atrazine; NA, not available; RIR, Renal ischemia-reperfusion; LYC, lycopene.

RCC

PANoptosis has been increasingly linked to tumorigenesis and cancer progression (12). The boundaries between different forms of PCD have become progressively less distinct, and therapeutic strategies targeting a single form of cell death often show limited efficacy (105). The discovery of PANoptosis not only broadens the understanding of cell death pathways but also offers novel approaches for cancer therapy. RCC, characterized by the proliferation of malignant epithelial cells, relies on strategies that effectively inhibit or eliminate these cells. Inducing PANoptosis, which simultaneously activates apoptosis, pyroptosis and necroptosis, holds potential therapeutic value for enhancing cancer cell clearance, and suppressing tumor growth and metastasis. PANoptosis has been investigated in several types of cancer, including colorectal cancer, breast cancer and low-grade glioma (12,128,129).

RCC is one of the most prevalent urological malignancies worldwide (130-132). Approximately one-third of patients present with metastatic RCC (mRCC) at the time of diagnosis, and the 5-year survival rate for mRCC remains at <30% (130,131). RCC is mainly classified into three histological subtypes: Clear cell RCC (ccRCC, also known as KIRC), papillary RCC (KIRP) and chromophobe RCC (KICH). Among these, KIRC is the most common subtype, accounting for ~70% of cases. As a highly immunogenic tumor, KIRC is treated using surgical resection and immune checkpoint inhibitors. Given its strong immunogenicity, PANoptosis may serve as a promising therapeutic target in KIRC (131,133-135).

Several studies have explored the association between PANoptosis and cancer prognosis using data from The Cancer Genome Atlas. One study reported that high expression of PANoptosis-related genes, such as AIM2, caspase-3, caspase-4 and TNFRSF10, was significantly associated with poor prognosis in KIRC (132). Conversely, in skin cutaneous melanoma, higher expression of genes including ZBP1, NLRP1, caspase-8 and GSDMD was shown to be associated with better survival outcomes (132). Further analysis has identified PANoptosis-related prognostic clusters, revealing underlying genetic mutations, distinct immune cell populations and activated oncogenic pathways that may account for differences in patient survival across tumor types (7). These findings highlight the heterogeneous nature of PANoptosis gene expression and suggest it may exert different roles depending on the cancer context.

Another study applied PANoptosis-related differentially expressed genes to predict prognosis in patients with KIRC. The high-risk PANoptosis group demonstrated an immunosuppressive tumor microenvironment, characterized by reduced infiltration of antitumor CD4+ T cells and natural killer cells, and elevated levels of immunosuppressive M2 macrophages and regulatory T cells. These findings suggest that PANoptosis may contribute to KIRC progression and influence immune regulation within the tumor microenvironment (9). In a separate study, Wang et al (133) developed a ccRCC prognostic risk model based on three specific PANoptosis-related microRNAs, revealing that patients in the high-risk group exhibited poorer outcomes and greater tumor malignancy. This group also showed enrichment of immune-related pathways and heightened immune activity, implying potential responsiveness to immunotherapy and chemotherapy. By contrast, the low-risk group demonstrated metabolic reprogramming, with increased fatty acid and amino acid metabolism. Furthermore, PANoptosis-related long non-coding RNAs have been associated with clinical outcomes, immune cell infiltration, immunosuppressive states, oncogenic signaling pathways and therapeutic responses in ccRCC. Single-cell sequencing identified LINC00944 as a potential biomarker linked to T-cell infiltration and PCD regulation, supporting its prognostic relevance (134). A recent study used bioinformatics analysis to characterize PANoptosis in the three RCC subtypes (KIRC, KIRP and KICH) and proposed a novel scoring system, the PANoptosis-Immunity Index (PANII). The PANII effectively reflected the immune microenvironment status across RCC subtypes and predicted response to immunotherapy. Functional validation revealed that knockout of the PYCARD gene significantly inhibited proliferation and invasion of KIRC cells in vitro, confirming its role in promoting tumor progression (135) (Table I).

CKD and DKD

CKD has emerged as a growing global public health concern, which markedly impairs patient quality of life. Over the past three decades, the global mortality rate associated with CKD has reached 41.5%, with ~1.2 million deaths reported in 2017. By 2040, CKD is projected to become the fifth leading cause of death worldwide (136,137). CKD is a progressive and long-term condition with diverse etiologies; however, renal fibrosis represents the common pathological hallmark driving progression to end-stage renal disease. The fibrotic process in CKD typically begins with the gradual loss of epithelial cells, including podocytes and tubular cells, as well as endothelial cells, which is followed by progressive glomerular sclerosis, collapse of peritubular capillaries and extensive tubulointerstitial fibrosis. The development of renal fibrosis is tightly associated with sustained inflammation and various forms of cell death (138-140). Regardless of the initial cause of kidney injury, nephrons are continuously lost, and fibrotic lesions advance until end-stage renal disease becomes inevitable. Therefore, targeting cell death pathways represents a potential therapeutic strategy to prevent or slow CKD progression by limiting the loss of functional kidney cells and suppressing inflammation. Notably, apoptosis, necroptosis and pyroptosis have been recognized as central pathogenic events in CKD of different etiologies. A large number of previous studies have confirmed that the pathological mechanisms of CKD and DKD are related to renal cell apoptosis (141-144), necroptosis (138,145-147) and pyroptosis (148,149).

Recent studies have revealed that three death modes occur simultaneously in CKD and DKD, which is different from the single death mode of PCD. Zhang et al (150) employed bioinformatics approaches to investigate the relationship between PANoptosis and CKD, identifying two central genes, FOS and PTGS2, as potential mediators. These genes have previously been associated with tissue inflammation, cell death and renal fibrosis, suggesting that PANoptosis may contribute to the onset and progression of CKD through their regulation (151-154). In a mouse model of post-burn CKD, elevated levels of caspase-1, caspase-3 and IL-1β have been observed in renal tissue, and apoptosis, pyroptosis and necroptosis were demonstrated to be concurrently activated (155). Sustained inflammation following burn injury led to caspase pathway activation, promoting PANoptosis, and aggravating renal injury and CKD progression. Treatment with the anti-inflammatory agent dexamethasone was able to suppress multiple forms of cell death and alleviate renal damage in this model (155) (Table I).

DKD, a major cause of end-stage kidney failure, is marked by progressive podocyte loss. Using single-cell RNA sequencing data, Lv et al (156) reported that TRAIL and death receptor 5 (DR5) were highly expressed in the podocytes of patients with DKD. These findings were further validated in renal biopsy samples. In vitro and in vivo experiments demonstrated that TRAIL induced the expression of PANoptosis-related markers, including caspase-1, caspase-3, phosphorylated (p)-MLKL and p-RIPK1, via DR5 in DKD podocytes. In addition, the simultaneous activation of apoptosis, pyroptosis and necroptosis exacerbated proteinuria and renal injury in mouse models of DKD. These effects were mitigated by TRAIL inhibition or gene knockout, indicating that the TRAIL/DR5 axis mediates podocyte PANoptosis injury in DKD (156) (Table I).

Collectively, these findings suggest that PANoptosis may serve as a promising therapeutic target in CKD and DKD. However, given the complex and heterogeneous nature of CKD pathogenesis, current research on PANoptosis in this context remains limited. Further investigation is needed to elucidate the mechanistic roles and therapeutic implications of PANoptosis across diverse CKD etiologies.

Renal injury caused by drugs and other factors

Recent studies have increasingly focused on the role of PANoptosis in drug-induced renal injury (6,10,11,157,158). One investigation revealed that the herbicide atrazine, known for its nephrotoxic effects, can downregulate Sam50 expression in the kidney, resulting in mitochondrial DNA (mtDNA) instability and the release of mtDNA into the cytoplasm. This mtDNA release activates the stimulator of IFN genes (STING) pathway, which subsequently upregulates inflammatory cytokines and key PANoptosis-associated molecules, including AIM2, ZBP1 and pyrin. These molecular events drive STING-dependent PANoptosis in renal tissue, ultimately leading to renal failure (10).

Triptolide-induced multi-organ damage has also been linked to a systemic inflammatory response and the activation of various lytic cell death pathways. Notably, the cell death triggered by triptolide cannot be mitigated by the inhibition of a single pathway. It has been shown that triptolide simultaneously activates markers of pyroptosis, apoptosis and necroptosis in macrophages. Immunofluorescence and immunoprecipitation assays have confirmed the colocalization and aggregation of ASC, RIPK3 and caspase-8, promoting the formation of the PANoptosome and inducing PANoptosis, which contributes to both nephrotoxicity and hepatotoxicity. Given that renal tubular epithelial cells and podocytes possess the molecular machinery required for all three forms of cell death, the authors of this previous study hypothesized that PANoptosis likely occurs in these renal cell types as well (157). In aristolochic acid nephropathy (AAN), administration of aristolochic acid I (AAI) has been shown to induce elevations in serum urea nitrogen and creatinine levels, and to induce PANoptosis in renal tubular epithelial cells. Both in vitro and in vivo experiments have demonstrated that inhibition of PANoptosis may alleviate AAI-induced renal injury, supporting its role in AAN pathogenesis (158). The anticancer agent CBL0137, either alone or in combination with LPS, has been shown to activate systemic inflammation and PANoptosis signaling. In mice, this treatment induced PANoptosis in macrophages and resulted in multi-organ injury, including kidney damage; mechanistically, CBL0137 promoted the formation of Z-DNA in macrophage nuclei, thereby activating ZBP1. By contrast, knockout of ZBP1 significantly reduced cell death induced by CBL0137 combined with LPS, highlighting the essential role of ZBP1 as a core component of the PANoptosome in mediating PANoptosis (11). In a mouse model sensitized to trichloroethylene, PANoptosis was observed in renal endothelial cells, and the pro-inflammatory cytokines TNFα and IFNγ appeared to act synergistically in driving this process. Inhibition of peripheral TNFα and IFNγ attenuated PANoptosis, preserved endothelial function and improved renal outcomes (6).

Beyond drug-induced renal injury, PANoptosis has also been implicated in renal damage resulting from non-chemical stressors. A recent study reported multi-organ injury, including damage to the kidneys and liver, following heatstroke. It was shown that ZBP1 mediates a PANoptosis-like cell death pathway during heatstroke, characterized by concurrent activation of apoptosis, pyroptosis and necroptosis. By contrast, deletion of ZBP1 abrogated this cell death process, and reversed the renal and hepatic damage induced by heatstroke (159) (Table I).

Treatment strategies and clinical significance of PANoptosis

Loss of parenchymal cells, together with proliferation or recruitment of maladaptive cells, disrupts cellular homeostasis and drives kidney injury and fibrosis (160,161). Modulating cell death programs may therefore represent an important approach to limiting renal damage across kidney diseases. Since PANoptosis encompasses multiple PCD patterns simultaneously, targeting and inhibiting these pathways in parallel may enhance renal protection. Although drugs directly targeting PANoptosis have not yet entered clinical trials, a number of compounds that act on its key node molecules are already in the clinical development stage for other indications (162-166), which provides a unique opportunity for the rapid initiation of clinical research on kidney diseases.

Molecules that act as central hubs in PANoptosis, such as ZBP1 and AIM2, can concurrently trigger pyroptosis, apoptosis and necroptosis in response to microbial or endogenous cues. Their inhibition or loss impairs activation of these effector modules and reduces cell death (82,167,168). Notably, these hubs have been implicated in AKI, RCC, drug-induced nephrotoxicity and other renal pathologies, highlighting their broad relevance. At present, specific inhibitors for ZBP1 and AIM2 are scarce, and traditional treatment strategies mainly focus on the inhibition of their downstream factors. However, some new technologies are attempting to precisely degrade ZBP1. For example, a recent study on the covalent recognition-based PROTAC molecule has confirmed that it can cause upregulation of ZBP1 expression in H1N1 infection models, inhibit the downstream necroptosis pathway, reduce the phosphorylation levels of RIPK3 and MLKL, and simultaneously lower the expression levels of IL-18, IL-6, IL-1β, TNF-α and IFN-β, effectively alleviating inflammation (169). In tumor research, it has been shown that the ZBP1 agonist CBL0137 can induce potent PANoptosis and antitumor immunity in mouse models of ZBP1-deficient melanoma (170). Conversely, necrosulfonamide, a small molecule inhibitor targeting the ZBP1 RHIM domain, can be used to alleviate therapeutic side effects (such as intestinal damage) (171). Recently, 4-sulfonic calixarenes have been described as being able to block the binding of dsDNA to the HIN200 domain of AIM2 and to inhibit AIM2 in vivo, further blocking the release of the downstream factors caspase-1, IL-1β and GSDMD (172). Further work revealed that these compounds are structurally similar to the clinically approved drug suramin, which can also inhibit AIM2 (172).

Therapeutic targeting of PANoptosome components is also considered promising. For example, the NLRP3 inhibitor MCC950 has been shown to attenuate kidney injury in models of AKI (173-175), CDK (176,177), DKD (178,179) and RCC (180). It has shown good safety and efficacy both in vivo and in vitro. Another NLRP3 inhibitor, OLT1177 (dapansutrile), has been proven to effectively reduce inflammatory indicators and has been shown to possess a good safety profile in a phase II trial of acute gout attacks (181). This also indicates prospects for the treatment of kidney diseases driven by metabolic stress, such as DKD. Z-VAD-FMK, a broad-spectrum caspase inhibitor widely used to suppress apoptosis (182), and Nec-1, a necroptosis inhibitor targeting RIPK1, have both demonstrated protective effects in various kidney diseases (182,183), but have not yet entered clinical trials. GSK2982772 is another RIPK1 inhibitor that has been tested for safety and efficacy in phase II clinical trials for psoriasis (165,166), rheumatoid arthritis (162) and ulcerative colitis (163). Given that RIPK1 is a key hub of PANoptosis in multiple nephropathy models, reusing it for the treatment of inflammatory nephropathy (such as lupus nephritis) is an attractive clinically available strategy. Caspase-8 functions as a key regulator across pathways, simultaneously influencing apoptosis, necroptosis and pyroptosis, thereby determining the cell death mode following activation signals (62). A recent study reported that inhibition of the caspase-8/caspase-3/NLRP3/GSDME-mediated pathway alleviated pyroptosis and rescued renal tubular cell injury induced by uric acid (184). Another investigation revealed that TP53INP2, an autophagy-related protein, can inhibit RCC progression by regulating the caspase-8 apoptotic pathway (185). Another caspase inhibitor (emricasan) has been shown in a clinical study on non-alcoholic steatohepatitis to significantly reduce ALT levels and the activation of caspase-3/7 in patients with non-alcoholic fatty liver disease. Although the results of this study have not been clinically translated, it provides a foundation for subsequent research on specific caspase inhibitors (such as those targeting caspase-8) (164). RIPK3 is also another important component of PANoptosomes. A novel inhibitor of RIPK3, Uh15-38, has been proven to effectively block necroptosis of alveolar epithelial cells caused by IAV infection in mouse models by inhibiting MLKL, and the levels of IL-1β and IL-18. In addition, it can reduce lung inflammation and mortality without affecting virus clearance or immune response (186). Its efficacy is consistent with other pre-clinical type I kinase inhibitors and comparable to that of currently Food and Drug Administration-approved drugs, such as sorafenib and dasatinib (186). In addition, sorafenib and edaravone have been shown to attenuate renal fibrosis induced by unilateral ureteral obstruction through inhibition of oxidative stress, inflammation and the RIPK3/MLKL pathway (187). Collectively, these findings suggest that targeting hub molecules and components of PANoptosis, including ZBP1, AIM2, RIPK1, NLRP3 and caspase family members, represents a promising therapeutic option for kidney diseases.

However, the interplay among apoptosis, pyroptosis and necroptosis complicates therapeutic development. These pathways often share molecular mediators and defense mechanisms, and they may coexist within the same kidney disease, acting either simultaneously or sequentially across different cell types. Furthermore, inhibition of one pathway may be compensated for by activation of others, resulting in limited therapeutic efficacy with single-target approaches. Therefore, identifying strategies that can simultaneously block multiple forms of cell death remains a key focus for future research.

Conclusion

Current evidence has indicated that PANoptosis may serve as a novel and promising therapeutic target in kidney diseases. Unlike interventions aimed at suppressing a single form of PCD, modulation of PANoptosis offers the potential to block multiple cell death pathways concurrently, thereby providing greater therapeutic benefit. This integrated approach represents a new direction for the prevention of kidney injury and the attenuation of disease progression; however, important challenges remain. Although key molecules involved in PANoptosis and their roles in distinct death pathways have been identified, the precise mechanisms by which these factors cooperate within the PANoptosome, and the hierarchy of control governing its assembly and signaling, are not yet fully understood. The dynamic and context-dependent composition of the PANoptosome, shaped by trigger signals, cell type and microenvironment, further complicates efforts to define universal assembly patterns. In vivo, notable technical barriers also exist in capturing transient, large multiprotein complexes and their intricate interaction networks. Additionally, the essential components, recruitment rules, sequential assembly mechanisms, and possible molecular switches or master regulators of PANoptosomes remain to be established. Finally, reports of PANoptosis in kidney diseases beyond AKI (8,110-112), CKD (150,155), DKD (156) and RCC (7,9,132-135) are limited (188). Its roles in hypertensive nephropathy, nephrotic syndrome, IgA nephropathy, lupus nephritis and polycystic kidney disease remain largely unexplored. These knowledge gaps highlight urgent priorities for future investigation.

Summary and outlook

Current research has revealed the mechanistic complexity of PANoptosis, and its involvement in the onset, progression and potential treatment of AKI, RCC, CKD, DKD and drug-induced renal injury. The activation and composition of the PANoptosome are highly variable, reflecting the context-dependent nature of PANoptosis and offering deeper insight into its regulatory role in cell death. As a result, PANoptosis has emerged as a key process closely associated with the development of various kidney diseases and presents a promising avenue for identifying novel therapeutic targets. Nevertheless, the role of PANoptosis in disease is not unidirectional; its effects on disease progression and treatment can be dual in nature, which is particularly evident in renal malignancies, where PANoptosis may exert both pro-tumor and antitumor effects. Furthermore, the dominant cell death pathways and the composition of PANoptotic complexes may vary depending on disease etiology or pathological stage. In AKI and CKD, greater attention should be directed toward cell death occurring within the renal parenchyma, whereas in RCC, further investigation is required to elucidate how the tumor immune microenvironment shapes PANoptosis. These observations underscore the need for continued in-depth research to elucidate its regulatory mechanisms and disease-specific outcomes. Although recent advances have expanded the understanding, the clinical application of PANoptosis in kidney disease remains in its early stages, and numerous questions remain unanswered. For example, while it is known that the composition of the PANoptosome varies with different stimuli or disease states, the mechanisms underlying this variability remain unclear. Additionally, although specific sensors are required to initiate PANoptosis in response to pathogens or damage-associated signals, the precise identity and disease-specific roles of these sensors remain to be fully defined. Therefore, several future research directions merit attention. First, it is essential to discover and validate reliable biomarkers that can facilitate the precise selection of therapeutic strategies targeting PANoptosis. Second, kidney cell-specific mechanisms should be elucidated, as tubular epithelial cells, podocytes and endothelial cells may assemble PANoptosomes with distinct components and functional outputs, which is crucial for therapeutic specificity. Third, a detailed characterization of the assembly and regulatory mechanisms of renal PANoptosomes is needed to support the identification of small-molecule targets. Fourth, across diverse renal diseases, the validation of hub molecules and the identification of regulators of PANoptosomes will provide a foundation for the development of selective inhibitors.

Since its initial conceptualization in 2019, the concept of PANoptosis has rapidly advanced as a field, offering a transformative framework to understand the intricate interplay between cell death and inflammation in kidney diseases. Unlike strategies that focus on blocking a single cell death pathway, targeting the integrated process of PANoptosis holds promise for achieving more effective protection of kidney tissue, attenuation of inflammation and improved clinical outcomes. Despite ongoing challenges, continued investigation into kidney-specific mechanisms of PANoptosis, together with the verification of therapeutic targets, the development of efficient and safe pharmacological agents, and the incorporation of precise biomarkers, is expected to drive major advances. Thus, targeted modulation of PANoptosis may emerge as a breakthrough approach in nephrology, providing novel therapeutic opportunities for currently refractory kidney diseases.

Availability of data and materials

Not applicable.

Authors' contributions

LJ wrote the original manuscript. YL and LZ revised the manuscript. ZF and MW searched the literature, and made substantial contributions to conception and design. ZM edited the manuscript. ML revised and supervised the manuscript. Data authentication is not applicable. All authors contributed to the article, and read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgements

Not applicable.

Funding

This work is funded by the National Nature Science Foundation of China (grant no. 82274482).

References

1 

Bedoui S, Herold MJ and Strasser A: Emerging connectivity of programmed cell death pathways and its physiological implications. Nat Rev Mol Cell Biol. 21:678–695. 2020. View Article : Google Scholar : PubMed/NCBI

2 

Newton K, Strasser A, Kayagaki N and Dixit VM: Cell death. Cell. 187:235–256. 2024. View Article : Google Scholar : PubMed/NCBI

3 

Malireddi RKS, Kesavardhana S and Kanneganti TD: ZBP1 and TAK1: Master regulators of NLRP3 inflammasome/pyroptosis, apoptosis, and necroptosis (PAN-optosis). Front Cell Infect Microbiol. 9:4062019. View Article : Google Scholar : PubMed/NCBI

4 

Pandian N and Kanneganti TD: PANoptosis: A unique innate immune inflammatory cell death modality. J Immunol. 209:1625–1633. 2022. View Article : Google Scholar : PubMed/NCBI

5 

Wang Y, Pandian N, Han JH, Sundaram B, Lee S, Karki R, Guy CS and Kanneganti TD: Single cell analysis of PANoptosome cell death complexes through an expansion microscopy method. Cell Mol Life Sci. 79:5312022. View Article : Google Scholar : PubMed/NCBI

6 

Xie H, Liang B, Zhu Q, Wang L, Li H, Qin Z, Zhang J, Liu Z and Wu Y: The role of PANoptosis in renal vascular endothelial cells: Implications for trichloroethylene-induced kidney injury. Ecotoxicol Environ Saf. 278:1164332024. View Article : Google Scholar : PubMed/NCBI

7 

Mall R and Kanneganti TD: Comparative analysis identifies genetic and molecular factors associated with prognostic clusters of PANoptosis in glioma, kidney and melanoma cancer. Sci Rep. 13:209622023. View Article : Google Scholar : PubMed/NCBI

8 

Uysal E, Dokur M, Kucukdurmaz F, Altınay S, Polat S, Batcıoglu K, Sezgın E, Sapmaz Erçakallı T, Yaylalı A, Yılmaztekin Y, et al: Targeting the PANoptosome with 3,4-meth ylenedioxy-β-nitrostyrene, reduces PANoptosis and protects the kidney against renal ischemia-reperfusion injury. J Invest Surg. 35:1824–1835. 2022. View Article : Google Scholar : PubMed/NCBI

9 

Jiang Z, Wang J, Dao C, Zhu M, Li Y, Liu F, Zhao Y, Li J, Yang Y and Pan Z: Utilizing a novel model of PANoptosis-related genes for enhanced prognosis and immune status prediction in kidney renal clear cell carcinoma. Apoptosis. 29:681–692. 2024. View Article : Google Scholar : PubMed/NCBI

10 

Yi BJ, Wang CC, Li XW, Xu YR, Ma XY, Jian PA, Talukder M, Li XN and Li JL: Lycopene protects against atrazine-induced kidney STING-dependent panoptosis through stabilizing mtDNA via interaction with Sam50/PHB1. J Agric Food Chem. 72:14956–14966. 2024. View Article : Google Scholar : PubMed/NCBI

11 

Li YP, Zhou ZY, Yan L, You YP, Ke HY, Yuan T, Yang HY, Xu R, Xu LH, Ouyang DY, et al: Inflammatory cell death PANoptosis is induced by the anti-cancer curaxin CBL0137 via eliciting the assembly of ZBP1-associated PANoptosome. Inflamm Res. 73:597–617. 2024. View Article : Google Scholar : PubMed/NCBI

12 

Karki R, Sharma BR, Lee E, Banoth B, Malireddi RKS, Samir P, Tuladhar S, Mummareddy H, Burton AR, Vogel P and Kanneganti TD: Interferon regulatory factor 1 regulates PANoptosis to prevent colorectal cancer. JCI Insight. 5:e1367202020. View Article : Google Scholar : PubMed/NCBI

13 

Kerr JF, Wyllie AH and Currie AR: Apoptosis: A basic biological phenomenon with wide-ranging implications in tissue kinetics. Br J Cancer. 26:239–257. 1972. View Article : Google Scholar : PubMed/NCBI

14 

Kroemer G: The pharmacology of T cell apoptosis. Adv Immunol. 58:211–296. 1995. View Article : Google Scholar : PubMed/NCBI

15 

Tsujimoto Y: Role of Bcl-2 family proteins in apoptosis: Apoptosomes or mitochondria? Genes Cells. 3:697–707. 1998. View Article : Google Scholar

16 

Li PF, Dietz R and von Harsdorf R: p53 regulates mitochondrial membrane potential through reactive oxygen species and induces cytochrome c-independent apoptosis blocked by Bcl-2. EMBO J. 18:6027–6036. 1999. View Article : Google Scholar : PubMed/NCBI

17 

Czabotar PE, Lessene G, Strasser A and Adams JM: Control of apoptosis by the BCL-2 protein family: Implications for physiology and therapy. Nat Rev Mol Cell Biol. 15:49–63. 2014. View Article : Google Scholar

18 

Kalkavan H and Green DR: MOMP, cell suicide as a BCL-2 family business. Cell Death Differ. 25:46–55. 2018. View Article : Google Scholar

19 

Yang J, Liu X, Bhalla K, Kim CN, Ibrado AM, Cai J, Peng TI, Jones DP and Wang X: Prevention of apoptosis by Bcl-2: Release of cytochrome c from mitochondria blocked. Science. 275:1129–1132. 1997. View Article : Google Scholar : PubMed/NCBI

20 

Green DR and Reed JC: Mitochondria and apoptosis. Science. 281:1309–1312. 1998. View Article : Google Scholar : PubMed/NCBI

21 

Hengartner MO and Horvitz HR: The ins and outs of programmed cell death during C. elegans development. Philos Trans R Soc Lond B Biol Sci. 345:243–246. 1994. View Article : Google Scholar : PubMed/NCBI

22 

Fox JL, Hughes MA, Meng X, Sarnowska NA, Powley IR, Jukes-Jones R, Dinsdale D, Ragan TJ, Fairall L, Schwabe JWR, et al: Cryo-EM structural analysis of FADD:Caspase-8 complexes defines the catalytic dimer architecture for co-ordinated control of cell fate. Nat Commun. 12:8192021. View Article : Google Scholar : PubMed/NCBI

23 

Fu TM, Li Y, Lu A, Li Z, Vajjhala PR, Cruz AC, Srivastava DB, DiMaio F, Penczek PA, Siegel RM, et al: Cryo-EM structure of caspase-8 tandem DED filament reveals assembly and regulation mechanisms of the death-inducing signaling complex. Mol Cell. 64:236–250. 2016. View Article : Google Scholar : PubMed/NCBI

24 

Feoktistova M, Geserick P, Kellert B, Dimitrova DP, Langlais C, Hupe M, Cain K, MacFarlane M, Häcker G and Leverkus M: cIAPs block Ripoptosome formation, a RIP1/caspase-8 containing intracellular cell death complex differentially regulated by cFLIP isoforms. Mol Cell. 43:449–463. 2011. View Article : Google Scholar : PubMed/NCBI

25 

Carneiro BA and El-Deiry WS: Targeting apoptosis in cancer therapy. Nat Rev Clin Oncol. 17:395–417. 2020. View Article : Google Scholar : PubMed/NCBI

26 

Van Opdenbosch N and Lamkanfi M: Caspases in cell death, inflammation, and disease. Immunity. 50:1352–1364. 2019. View Article : Google Scholar : PubMed/NCBI

27 

Seol DW, Li J, Seol MH, Park SY, Talanian RV and Billiar TR: Signaling events triggered by tumor necrosis factor-related apoptosis-inducing ligand (TRAIL): Caspase-8 is required for TRAIL-induced apoptosis. Cancer Res. 61:1138–1143. 2001.PubMed/NCBI

28 

Degterev A, Huang Z, Boyce M, Li Y, Jagtap P, Mizushima N, Cuny GD, Mitchison TJ, Moskowitz MA and Yuan J: Chemical inhibitor of nonapoptotic cell death with therapeutic potential for ischemic brain injury. Nat Chem Biol. 1:112–119. 2005. View Article : Google Scholar

29 

Khoury MK, Gupta K, Franco SR and Liu B: Necroptosis in the pathophysiology of disease. Am J Pathol. 190:272–285. 2020. View Article : Google Scholar :

30 

Weinlich R, Oberst A, Beere HM and Green DR: Necroptosis in development, inflammation and disease. Nat Rev Mol Cell Biol. 18:127–136. 2017. View Article : Google Scholar

31 

Murphy JM: The killer pseudokinase mixed lineage kinase domain-like protein (MLKL). Cold Spring Harb Perspect Biol. 12:a0363762020. View Article : Google Scholar

32 

Petrie EJ, Sandow JJ, Lehmann WIL, Liang LY, Coursier D, Young SN, Kersten WJA, Fitzgibbon C, Samson AL, Jacobsen AV, et al: Viral MLKL homologs subvert necroptotic cell death by sequestering cellular RIPK3. Cell Rep. 28:3309–3319.e5. 2019. View Article : Google Scholar : PubMed/NCBI

33 

Holler N, Zaru R, Micheau O, Thome M, Attinger A, Valitutti S, Bodmer JL, Schneider P, Seed B and Tschopp J: Fas triggers an alternative, caspase-8-independent cell death pathway using the kinase RIP as effector molecule. Nat Immunol. 1:489–495. 2000. View Article : Google Scholar

34 

He S, Liang Y, Shao F and Wang X: Toll-like receptors activate programmed necrosis in macrophages through a receptor-interacting kinase-3-mediated pathway. Proc Natl Acad Sci USA. 108:20054–20059. 2011. View Article : Google Scholar : PubMed/NCBI

35 

Kaiser WJ, Sridharan H, Huang C, Mandal P, Upton JW, Gough PJ, Sehon CA, Marquis RW, Bertin J and Mocarski ES: Toll-like receptor 3-mediated necrosis via TRIF, RIP3, and MLKL. J Biol Chem. 288:31268–31279. 2013. View Article : Google Scholar : PubMed/NCBI

36 

Zhang T, Wang Y, Inuzuka H and Wei W: Necroptosis pathways in tumorigenesis. Semin Cancer Biol. 86:32–40. 2022. View Article : Google Scholar : PubMed/NCBI

37 

Cho YS, Challa S, Moquin D, Genga R, Ray TD, Guildford M and Chan FK: Phosphorylation-driven assembly of the RIP1-RIP3 complex regulates programmed necrosis and virus-induced inflammation. Cell. 137:1112–1123. 2009. View Article : Google Scholar : PubMed/NCBI

38 

Vanden Berghe T, Kaiser WJ, Bertrand MJ and Vandenabeele P: Molecular crosstalk between apoptosis, necroptosis, and survival signaling. Mol Cell Oncol. 2:e9750932015. View Article : Google Scholar

39 

Wong WW, Gentle IE, Nachbur U, Anderton H, Vaux DL and Silke J: RIPK1 is not essential for TNFR1-induced activation of NF-kappaB. Cell Death Differ. 17:482–487. 2010. View Article : Google Scholar

40 

Wang H, Sun L, Su L, Rizo J, Liu L, Wang LF, Wang FS and Wang X: Mixed lineage kinase domain-like protein MLKL causes necrotic membrane disruption upon phosphorylation by RIP3. Mol Cell. 54:133–146. 2014. View Article : Google Scholar : PubMed/NCBI

41 

Cai Z, Jitkaew S, Zhao J, Chiang HC, Choksi S, Liu J, Ward Y, Wu LG and Liu ZG: Plasma membrane translocation of trimerized MLKL protein is required for TNF-induced necroptosis. Nat Cell Biol. 16:55–65. 2014. View Article : Google Scholar

42 

Robinson N, McComb S, Mulligan R, Dudani R, Krishnan L and Sad S: Type I interferon induces necroptosis in macrophages during infection with Salmonella enterica serovar Typhimurium. Nat Immunol. 13:954–962. 2012. View Article : Google Scholar : PubMed/NCBI

43 

McComb S, Cessford E, Alturki NA, Joseph J, Shutinoski B, Startek JB, Gamero AM, Mossman KL and Sad S: Type-I interferon signaling through ISGF3 complex is required for sustained Rip3 activation and necroptosis in macrophages. Proc Natl Acad Sci USA. 111:E3206–E3213. 2014. View Article : Google Scholar : PubMed/NCBI

44 

Xia B, Fang S, Chen X, Hu H, Chen P, Wang H and Gao Z: MLKL forms cation channels. Cell Res. 26:517–528. 2016. View Article : Google Scholar : PubMed/NCBI

45 

Negroni A, Colantoni E, Cucchiara S and Stronati L: Necroptosis in intestinal inflammation and cancer: New concepts and therapeutic perspectives. Biomolecules. 10:14312020. View Article : Google Scholar : PubMed/NCBI

46 

Woo Y, Lee HJ, Jung YM and Jung YJ: Regulated necrotic cell death in alternative tumor therapeutic strategies. Cells. 9:27092020. View Article : Google Scholar : PubMed/NCBI

47 

Zychlinsky A, Prevost MC and Sansonetti PJ: Shigella flexneri induces apoptosis in infected macrophages. Nature. 358:167–169. 1992. View Article : Google Scholar : PubMed/NCBI

48 

Hilbi H, Moss JE, Hersh D, Chen Y, Arondel J, Banerjee S, Flavell RA, Yuan J, Sansonetti PJ and Zychlinsky A: Shigella-induced apoptosis is dependent on caspase-1 which binds to IpaB. J Biol Chem. 273:32895–32900. 1998. View Article : Google Scholar : PubMed/NCBI

49 

Cookson BT and Brennan MA: Pro-inflammatory programmed cell death. Trends Microbiol. 9:113–114. 2001. View Article : Google Scholar : PubMed/NCBI

50 

Wallach D, Kang TB, Dillon CP and Green DR: Programmed necrosis in inflammation: Toward identification of the effector molecules. Science. 352:aaf21542016. View Article : Google Scholar : PubMed/NCBI

51 

Liu X, Zhang Z, Ruan J, Pan Y, Magupalli VG, Wu H and Lieberman J: Inflammasome-activated gasdermin D causes pyroptosis by forming membrane pores. Nature. 535:153–158. 2016. View Article : Google Scholar : PubMed/NCBI

52 

Atabaki R, Khaleghzadeh-Ahangar H, Esmaeili N and Mohseni-Moghaddam P: Role of pyroptosis, a pro-inflammatory programmed cell death, in epilepsy. Cell Mol Neurobiol. 43:1049–1059. 2023. View Article : Google Scholar

53 

Bergsbaken T, Fink SL and Cookson BT: Pyroptosis: Host cell death and inflammation. Nat Rev Microbiol. 7:99–109. 2009. View Article : Google Scholar : PubMed/NCBI

54 

Rathinam VAK, Vanaja SK, Waggoner L, Sokolovska A, Becker C, Stuart LM, Leong JM and Fitzgerald KA: TRIF licenses caspase-11-dependent NLRP3 inflammasome activation by gram-negative bacteria. Cell. 150:606–619. 2012. View Article : Google Scholar : PubMed/NCBI

55 

Fritsch M, Günther SD, Schwarzer R, Albert MC, Schorn F, Werthenbach JP, Schiffmann LM, Stair N, Stocks H, Seeger JM, et al: Caspase-8 is the molecular switch for apoptosis, necroptosis and pyroptosis. Nature. 575:683–687. 2019. View Article : Google Scholar : PubMed/NCBI

56 

Gurung P, Anand PK, Malireddi RK, Vande Walle L, Van Opdenbosch N, Dillon CP, Weinlich R, Green DR, Lamkanfi M and Kanneganti TD: FADD and caspase-8 mediate priming and activation of the canonical and noncanonical Nlrp3 inflammasomes. J Immunol. 192:1835–1846. 2014. View Article : Google Scholar : PubMed/NCBI

57 

Man SM, Tourlomousis P, Hopkins L, Monie TP, Fitzgerald KA and Bryant CE: Salmonella infection induces recruitment of caspase-8 to the inflammasome to modulate IL-1β production. J Immunol. 191:5239–5246. 2013. View Article : Google Scholar : PubMed/NCBI

58 

Van Opdenbosch N, Van Gorp H, Verdonckt M, Saavedra PHV, de Vasconcelos NM, Gonçalves A, Vande Walle L, Demon D, Matusiak M, Van Hauwermeiren F, et al: Caspase-1 engagement and TLR-induced c-FLIP expression suppress ASC/caspase-8-dependent apoptosis by inflammasome sensors NLRP1b and NLRC4. Cell Rep. 21:3427–3444. 2017. View Article : Google Scholar : PubMed/NCBI

59 

Park MY, Ha SE, Vetrivel P, Kim HH, Bhosale PB, Abusaliya A and Kim GS: Differences of key proteins between apoptosis and necroptosis. Biomed Res Int. 2021:34201682021. View Article : Google Scholar : PubMed/NCBI

60 

Newton K, Wickliffe KE, Dugger DL, Maltzman A, Roose-Girma M, Dohse M, Kőműves L, Webster JD and Dixit VM: Cleavage of RIPK1 by caspase-8 is crucial for limiting apoptosis and necroptosis. Nature. 574:428–431. 2019. View Article : Google Scholar : PubMed/NCBI

61 

Vandenabeele P, Galluzzi L, Vanden Berghe T and Kroemer G: Molecular mechanisms of necroptosis: An ordered cellular explosion. Nat Rev Mol Cell Biol. 11:700–714. 2010. View Article : Google Scholar : PubMed/NCBI

62 

Schwarzer R, Laurien L and Pasparakis M: New insights into the regulation of apoptosis, necroptosis, and pyroptosis by receptor interacting protein kinase 1 and caspase-8. Curr Opin Cell Biol. 63:186–193. 2020. View Article : Google Scholar : PubMed/NCBI

63 

Hsu H, Shu HB, Pan MG and Goeddel DV: TRADD-TRAF2 and TRADD-FADD interactions define two distinct TNF receptor 1 signal transduction pathways. Cell. 84:299–308. 1996. View Article : Google Scholar : PubMed/NCBI

64 

Dillon CP, Weinlich R, Rodriguez DA, Cripps JG, Quarato G, Gurung P, Verbist KC, Brewer TL, Llambi F, Gong YN, et al: RIPK1 blocks early postnatal lethality mediated by caspase-8 and RIPK3. Cell. 157:1189–1202. 2014. View Article : Google Scholar : PubMed/NCBI

65 

Orning P, Weng D, Starheim K, Ratner D, Best Z, Lee B, Brooks A, Xia S, Wu H, Kelliher MA, et al: Pathogen blockade of TAK1 triggers caspase-8-dependent cleavage of gasdermin D and cell death. Science. 362:1064–1069. 2018. View Article : Google Scholar : PubMed/NCBI

66 

Sarhan J, Liu BC, Muendlein HI, Li P, Nilson R, Tang AY, Rongvaux A, Bunnell SC, Shao F, Green DR and Poltorak A: Caspase-8 induces cleavage of gasdermin D to elicit pyroptosis during Yersinia infection. Proc Natl Acad Sci USA. 115:E10888–E10897. 2018. View Article : Google Scholar : PubMed/NCBI

67 

Hou J, Zhao R, Xia W, Chang CW, You Y, Hsu JM, Nie L, Chen Y, Wang YC, Liu C, et al: PD-L1-mediated gasdermin C expression switches apoptosis to pyroptosis in cancer cells and facilitates tumour necrosis. Nat Cell Biol. 22:1264–1275. 2020. View Article : Google Scholar : PubMed/NCBI

68 

Wang Y, Karki R, Zheng M, Kancharana B, Lee S, Kesavardhana S, Hansen BS, Pruett-Miller SM and Kanneganti TD: Cutting edge: Caspase-8 is a linchpin in caspase-3 and gasdermin D activation to control cell death, cytokine release, and host defense during influenza A virus infection. J Immunol. 207:2411–2416. 2021. View Article : Google Scholar : PubMed/NCBI

69 

Rogers C, Fernandes-Alnemri T, Mayes L, Alnemri D, Cingolani G and Alnemri ES: Cleavage of DFNA5 by caspase-3 during apoptosis mediates progression to secondary necrotic/pyroptotic cell death. Nat Commun. 8:141282017. View Article : Google Scholar : PubMed/NCBI

70 

Wang Y, Gao W, Shi X, Ding J, Liu W, He H, Wang K and Shao F: Chemotherapy drugs induce pyroptosis through caspase-3 cleavage of a gasdermin. Nature. 547:99–103. 2017. View Article : Google Scholar : PubMed/NCBI

71 

Tsuchiya K, Nakajima S, Hosojima S, Thi Nguyen D, Hattori T, Manh Le T, Hori O, Mahib MR, Yamaguchi Y, Miura M, et al: Caspase-1 initiates apoptosis in the absence of gasdermin D. Nat Commun. 10:20912019. View Article : Google Scholar : PubMed/NCBI

72 

Lamkanfi M, Kanneganti TD, Van Damme P, Vanden Berghe T, Vanoverberghe I, Vandekerckhove J, Vandenabeele P, Gevaert K and Núñez G: Targeted peptidecentric proteomics reveals caspase-7 as a substrate of the caspase-1 inflammasomes. Mol Cell Proteomics. 7:2350–2363. 2008. View Article : Google Scholar : PubMed/NCBI

73 

Conos SA, Chen KW, De Nardo D, Hara H, Whitehead L, Núñez G, Masters SL, Murphy JM, Schroder K, Vaux DL, et al: Active MLKL triggers the NLRP3 inflammasome in a cell-intrinsic manner. Proc Natl Acad Sci USA. 114:E961–E969. 2017. View Article : Google Scholar : PubMed/NCBI

74 

Gutierrez KD, Davis MA, Daniels BP, Olsen TM, Ralli-Jain P, Tait SW, Gale M Jr and Oberst A: MLKL activation triggers NLRP3-mediated processing and release of IL-1β independently of gasdermin-D. J Immunol. 198:2156–2164. 2017. View Article : Google Scholar : PubMed/NCBI

75 

Muñoz-Planillo R, Kuffa P, Martínez-Colón G, Smith BL, Rajendiran TM and Núñez G: K+ efflux is the common trigger of NLRP3 inflammasome activation by bacterial toxins and particulate matter. Immunity. 38:1142–1153. 2013. View Article : Google Scholar

76 

Lawlor KE, Khan N, Mildenhall A, Gerlic M, Croker BA, D'Cruz AA, Hall C, Kaur Spall S, Anderton H, Masters SL, et al: RIPK3 promotes cell death and NLRP3 inflammasome activation in the absence of MLKL. Nat Commun. 6:62822015. View Article : Google Scholar : PubMed/NCBI

77 

Malireddi RKS, Gurung P, Mavuluri J, Dasari TK, Klco JM, Chi H and Kanneganti TD: TAK1 restricts spontaneous NLRP3 activation and cell death to control myeloid proliferation. J Exp Med. 215:1023–1034. 2018. View Article : Google Scholar : PubMed/NCBI

78 

Zheng M, Karki R, Vogel P and Kanneganti TD: Caspase-6 is a key regulator of innate immunity, inflammasome activation, and host defense. Cell. 181:674–687.e13. 2020. View Article : Google Scholar : PubMed/NCBI

79 

Malireddi RKS, Gurung P, Kesavardhana S, Samir P, Burton A, Mummareddy H, Vogel P, Pelletier S, Burgula S and Kanneganti TD: Innate immune priming in the absence of TAK1 drives RIPK1 kinase activity-independent pyroptosis, apoptosis, necroptosis, and inflammatory disease. J Exp Med. 217:jem.201916442020. View Article : Google Scholar

80 

Christgen S, Tweedell RE and Kanneganti TD: Programming inflammatory cell death for therapy. Pharmacol Ther. 232:1080102022. View Article : Google Scholar :

81 

Tweedell RE and Kanneganti TD: Advances in inflammasome research: Recent breakthroughs and future hurdles. Trends Mol Med. 26:969–971. 2020. View Article : Google Scholar : PubMed/NCBI

82 

Lee S, Karki R, Wang Y, Nguyen LN, Kalathur RC and Kanneganti TD: AIM2 forms a complex with pyrin and ZBP1 to drive PANoptosis and host defence. Nature. 597:415–419. 2021. View Article : Google Scholar : PubMed/NCBI

83 

Shi C, Cao P, Wang Y, Zhang Q, Zhang D, Wang Y, Wang L and Gong Z: PANoptosis: A cell death characterized by pyroptosis, apoptosis, and necroptosis. J Inflamm Res. 16:1523–1532. 2023. View Article : Google Scholar : PubMed/NCBI

84 

Banoth B, Tuladhar S, Karki R, Sharma BR, Briard B, Kesavardhana S, Burton A and Kanneganti TD: ZBP1 promotes fungi-induced inflammasome activation and pyroptosis, apoptosis, and necroptosis (PANoptosis). J Biol Chem. 295:18276–18283. 2020. View Article : Google Scholar : PubMed/NCBI

85 

Kesavardhana S, Malireddi RKS, Burton AR, Porter SN, Vogel P, Pruett-Miller SM and Kanneganti TD: The Zα2 domain of ZBP1 is a molecular switch regulating influenza-induced PANoptosis and perinatal lethality during development. J Biol Chem. 295:8325–8330. 2020. View Article : Google Scholar : PubMed/NCBI

86 

Karki R, Lee S, Mall R, Pandian N, Wang Y, Sharma BR, Malireddi RS, Yang D, Trifkovic S, Steele JA, et al: ZBP1-dependent inflammatory cell death, PANoptosis, and cytokine storm disrupt IFN therapeutic efficacy during coronavirus infection. Sci Immunol. 7:eabo62942022. View Article : Google Scholar : PubMed/NCBI

87 

Malireddi RKS, Kesavardhana S, Karki R, Kancharana B, Burton AR and Kanneganti TD: RIPK1 distinctly regulates Yersinia-induced inflammatory cell death, PANoptosis. Immunohorizons. 4:789–796. 2020. View Article : Google Scholar : PubMed/NCBI

88 

Sundaram B, Pandian N, Mall R, Wang Y, Sarkar R, Kim HJ, Malireddi RKS, Karki R, Janke LJ, Vogel P and Kanneganti TD: NLRP12-PANoptosome activates PANoptosis and pathology in response to heme and PAMPs. Cell. 186:2783–2801.e20. 2023. View Article : Google Scholar : PubMed/NCBI

89 

Kuriakose T, Man SM, Malireddi RK, Karki R, Kesavardhana S, Place DE, Neale G, Vogel P and Kanneganti TD: ZBP1/DAI is an innate sensor of influenza virus triggering the NLRP3 inflammasome and programmed cell death pathways. Sci Immunol. 1:aag20452016. View Article : Google Scholar : PubMed/NCBI

90 

Rebsamen M, Heinz LX, Meylan E, Michallet MC, Schroder K, Hofmann K, Vazquez J, Benedict CA and Tschopp J: DAI/ZBP1 recruits RIP1 and RIP3 through RIP homotypic interaction motifs to activate NF-kappaB. EMBO Rep. 10:916–922. 2009. View Article : Google Scholar : PubMed/NCBI

91 

Upton JW, Kaiser WJ and Mocarski ES: DAI/ZBP1/DLM-1 complexes with RIP3 to mediate virus-induced programmed necrosis that is targeted by murine cytomegalovirus vIRA. Cell Host Microbe. 11:290–297. 2012. View Article : Google Scholar : PubMed/NCBI

92 

Kuriakose T and Kanneganti TD: ZBP1: Innate sensor regulating cell death and inflammation. Trends Immunol. 39:123–134. 2018. View Article : Google Scholar :

93 

Thapa RJ, Ingram JP, Ragan KB, Nogusa S, Boyd DF, Benitez AA, Sridharan H, Kosoff R, Shubina M, Landsteiner VJ, et al: DAI senses influenza A virus genomic RNA and activates RIPK3-dependent cell death. Cell Host Microbe. 20:674–681. 2016. View Article : Google Scholar : PubMed/NCBI

94 

Zheng M and Kanneganti TD: Newly identified function of caspase-6 in ZBP1-mediated innate immune responses, NLRP3 inflammasome activation, PANoptosis, and host defense. J Cell Immunol. 2:341–347. 2020.

95 

Momota M, Lelliott P, Kubo A, Kusakabe T, Kobiyama K, Kuroda E, Imai Y, Akira S, Coban C and Ishii KJ: ZBP1 governs the inflammasome-independent IL-1α and neutrophil inflammation that play a dual role in anti-influenza virus immunity. Int Immunol. 32:203–212. 2020. View Article : Google Scholar

96 

Hornung V, Ablasser A, Charrel-Dennis M, Bauernfeind F, Horvath G, Caffrey DR, Latz E and Fitzgerald KA: AIM2 recognizes cytosolic dsDNA and forms a caspase-1-activating inflammasome with ASC. Nature. 458:514–518. 2009. View Article : Google Scholar : PubMed/NCBI

97 

Bürckstümmer T, Baumann C, Blüml S, Dixit E, Dürnberger G, Jahn H, Planyavsky M, Bilban M, Colinge J, Bennett KL and Superti-Furga G: An orthogonal proteomic-genomic screen identifies AIM2 as a cytoplasmic DNA sensor for the inflammasome. Nat Immunol. 10:266–272. 2009. View Article : Google Scholar : PubMed/NCBI

98 

Fernandes-Alnemri T, Yu JW, Datta P, Wu J and Alnemri ES: AIM2 activates the inflammasome and cell death in response to cytoplasmic DNA. Nature. 458:509–513. 2009. View Article : Google Scholar : PubMed/NCBI

99 

Fernandes-Alnemri T, Yu JW, Juliana C, Solorzano L, Kang S, Wu J, Datta P, McCormick M, Huang L, McDermott E, et al: The AIM2 inflammasome is critical for innate immunity to Francisella tularensis. Nat Immunol. 11:385–393. 2010. View Article : Google Scholar : PubMed/NCBI

100 

Rathinam VA, Jiang Z, Waggoner SN, Sharma S, Cole LE, Waggoner L, Vanaja SK, Monks BG, Ganesan S, Latz E, et al: The AIM2 inflammasome is essential for host defense against cytosolic bacteria and DNA viruses. Nat Immunol. 11:395–402. 2010. View Article : Google Scholar : PubMed/NCBI

101 

Vanden Berghe T, Linkermann A, Jouan-Lanhouet S, Walczak H and Vandenabeele P: Regulated necrosis: The expanding network of non-apoptotic cell death pathways. Nat Rev Mol Cell Biol. 15:135–147. 2014. View Article : Google Scholar : PubMed/NCBI

102 

Kist M, Kőműves LG, Goncharov T, Dugger DL, Yu C, Roose-Girma M, Newton K, Webster JD and Vucic D: Impaired RIPK1 ubiquitination sensitizes mice to TNF toxicity and inflammatory cell death. Cell Death Differ. 28:985–1000. 2021. View Article : Google Scholar :

103 

Mihaly SR, Ninomiya-Tsuji J and Morioka S: TAK1 control of cell death. Cell Death Differ. 21:1667–1676. 2014. View Article : Google Scholar : PubMed/NCBI

104 

Sanjo H, Nakayama J, Yoshizawa T, Fehling HJ, Akira S and Taki S: Cutting edge: TAK1 safeguards macrophages against proinflammatory cell death. J Immunol. 203:783–788. 2019. View Article : Google Scholar : PubMed/NCBI

105 

Malireddi RKS, Tweedell RE and Kanneganti TD: PANoptosis components, regulation, and implications. Aging (Albany NY). 12:11163–11164. 2020. View Article : Google Scholar : PubMed/NCBI

106 

Kanneganti TD: Intracellular innate immune receptors: Life inside the cell. Immunol Rev. 297:5–12. 2020. View Article : Google Scholar : PubMed/NCBI

107 

Ostermann M, Basu RK and Mehta RL: Acute kidney injury. Intensive Care Med. 49:219–222. 2023. View Article : Google Scholar : PubMed/NCBI

108 

Ostermann M, Lumlertgul N, Jeong R, See E, Joannidis M and James M: Acute kidney injury. Lancet. 405:241–256. 2025. View Article : Google Scholar : PubMed/NCBI

109 

Wu C, Zhang Y, Nie S, Hong D, Zhu J, Chen Z, Liu B, Liu H, Yang Q, Li H, et al: Predicting in-hospital outcomes of patients with acute kidney injury. Nat Commun. 14:37392023. View Article : Google Scholar : PubMed/NCBI

110 

Wei S, Wu L, Xiang Z, Yang X, Pei D, Jiang L and Du Z: EIF2AK2 protein targeted activation of AIM2-mediated PANoptosis promotes sepsis-induced acute kidney injury. Ren Fail. 46:24036492024. View Article : Google Scholar : PubMed/NCBI

111 

Lin SY, Chang CL, Liou KT, Kao YK, Wang YH, Chang CC, Kuo TBJ, Huang HT, Yang CCH, Liaw CC and Shen YC: The protective role of Achyranthes aspera extract against cisplatininduced nephrotoxicity by alleviating oxidative stress, inflammation, and PANoptosis. J Ethnopharmacol. 319:1170972024. View Article : Google Scholar

112 

You R: Mining biomarkers of acute kidney injury in sepsis and preliminary exploration of the PANoptosis-related mechanism. PhD dissertation. Peking Union Medical College; 2023

113 

Yan WT, Zhao WJ, Hu XM, Ban XX, Ning WY, Wan H, Zhang Q and Xiong K: PANoptosis-like cell death in ischemia/reperfusion injury of retinal neurons. Neural Regen Res. 18:357–363. 2023.

114 

Wan H, Ban X, He Y, Yang Y, Hu X, Shang L, Wan X, Zhang Q and Xiong K: Voltage-dependent anion channel 1 oligomerization regulates PANoptosis in retinal ischemia-reperfusion injury. Neural Regen Res. 21:1652–1664. 2026. View Article : Google Scholar

115 

Yan WT, Yang YD, Hu XM, Ning WY, Liao LS, Lu S, Zhao WJ, Zhang Q and Xiong K: Do pyroptosis, apoptosis, and necroptosis (PANoptosis) exist in cerebral ischemia? Evidence from cell and rodent studies. Neural Regen Res. 17:1761–1768. 2022. View Article : Google Scholar : PubMed/NCBI

116 

Hamar P, Song E, Kökény G, Chen A, Ouyang N and Lieberman J: Small interfering RNA targeting Fas protects mice against renal ischemia-reperfusion injury. Proc Natl Acad Sci USA. 101:14883–14888. 2004. View Article : Google Scholar : PubMed/NCBI

117 

Zheng X, Zang G, Jiang J, He W, Johnston NJ, Ling H, Chen R, Zhang X, Liu Y, Haig A, et al: Attenuating ischemia-reperfusion injury in kidney transplantation by perfusing donor organs with siRNA cocktail solution. Transplantation. 100:743–752. 2016. View Article : Google Scholar : PubMed/NCBI

118 

Basile DP, Liapis H and Hammerman MR: Expression of bcl-2 and bax in regenerating rat renal tubules following ischemic injury. Am J Physiol. 272:F640–F647. 1997.PubMed/NCBI

119 

Thomas K, Zondler L, Ludwig N, Kardell M, Lüneburg C, Henke K, Mersmann S, Margraf A, Spieker T, Tekath T, et al: Glutamine prevents acute kidney injury by modulating oxidative stress and apoptosis in tubular epithelial cells. JCI Insight. 7:e1631612022. View Article : Google Scholar : PubMed/NCBI

120 

Yang B, Wang J, Qiao J, Zhang Q, Liu Q, Tan Y, Wang Q, Sun W, Feng W, Li Z, et al: Circ DENND4C inhibits pyroptosis and alleviates ischemia-reperfusion acute kidney injury by exosomes secreted from human urine-derived stem cells. Chem Biol Interact. 391:1109222024. View Article : Google Scholar : PubMed/NCBI

121 

Tonnus W, Maremonti F, Belavgeni A, Latk M, Kusunoki Y, Brucker A, von Mässenhausen A, Meyer C, Locke S, Gembardt F, et al: Gasdermin D-deficient mice are hypersensitive to acute kidney injury. Cell Death Dis. 13:7922022. View Article : Google Scholar : PubMed/NCBI

122 

Miao N, Yin F, Xie H, Wang Y, Xu Y, Shen Y, Xu D, Yin J, Wang B, Zhou Z, et al: The cleavage of gasdermin D by caspase-11 promotes tubular epithelial cell pyroptosis and urinary IL-18 excretion in acute kidney injury. Kidney Int. 96:1105–1120. 2019. View Article : Google Scholar : PubMed/NCBI

123 

Li T, Sun H, Li Y, Su L, Jiang J, Liu Y, Jiang N, Huang R, Zhang J and Peng Z: Downregulation of macrophage migration inhibitory factor attenuates NLRP3 inflammasome mediated pyroptosis in sepsis-induced AKI. Cell Death Discov. 8:612022. View Article : Google Scholar : PubMed/NCBI

124 

Linkermann A and Green DR: Necroptosis. N Engl J Med. 370:455–465. 2014. View Article : Google Scholar : PubMed/NCBI

125 

Linkermann A, Bräsen JH, Himmerkus N, Liu S, Huber TB, Kunzendorf U and Krautwald S: Rip1 (receptor-interacting protein kinase 1) mediates necroptosis and contributes to renal ischemia/reperfusion injury. Kidney Int. 81:751–761. 2012. View Article : Google Scholar : PubMed/NCBI

126 

Wang JN, Liu MM, Wang F, Wei B, Yang Q, Cai YT, Chen X, Liu XQ, Jiang L, Li C, et al: RIPK1 inhibitor Cpd-71 attenuates renal dysfunction in cisplatin-treated mice via attenuating necroptosis, inflammation and oxidative stress. Clin Sci (Lond). 133:1609–1627. 2019. View Article : Google Scholar : PubMed/NCBI

127 

Chen C, Xie J, Chen Z, Ye K, Wu C, Dai X, Yuan Y, Lin Y, Wang Y, Chen H, et al: Role of Z-DNA binding protein 1 sensing mitochondrial Z-DNA and triggering necroptosis in oxalate-induced acute kidney injury. J Am Soc Nephrol. 36:361–377. 2025. View Article : Google Scholar

128 

He P, Ma Y, Wu Y, Zhou Q and Du H: Exploring PANoptosis in breast cancer based on scRNA-seq and bulk-seq. Front Endocrinol (Lausanne). 14:11649302023. View Article : Google Scholar : PubMed/NCBI

129 

Chen G, He Z, Jiang W, Li L, Luo B, Wang X and Zheng X: Construction of a machine learning-based artificial neural network for discriminating PANoptosis related subgroups to predict prognosis in low-grade gliomas. Sci Rep. 12:221192022. View Article : Google Scholar : PubMed/NCBI

130 

Liu W, Liu Y, Chen S, Hui J and He S: AURKB promotes immunogenicity and immune infiltration in clear cell renal cell carcinoma. Discov Oncol. 15:2862024. View Article : Google Scholar : PubMed/NCBI

131 

Choueiri TK and Motzer RJ: Systemic therapy for metastatic renal-cell carcinoma. N Engl J Med. 376:354–366. 2017. View Article : Google Scholar : PubMed/NCBI

132 

Mall R, Bynigeri RR, Karki R, Malireddi RKS, Sharma BR and Kanneganti TD: Pancancer transcriptomic profiling identifies key PANoptosis markers as therapeutic targets for oncology. NAR Cancer. 4:zcac0332022. View Article : Google Scholar : PubMed/NCBI

133 

Wang Y, Zhou J, Zhang N, Zhu Y, Zhong Y, Wang Z, Jin H and Wang X: A novel defined PANoptosis-related miRNA signature for predicting the prognosis and immune characteristics in clear cell renal cell carcinoma: A miRNA signature for the prognosis of ccRCC. Int J Mol Sci. 24:93922023. View Article : Google Scholar : PubMed/NCBI

134 

Liu W, Qu C and Wang X: Comprehensive analysis of the role of immune-related PANoptosis lncRNA model in renal clear cell carcinoma based on RNA transcriptome and single-cell sequencing. Oncol Res. 31:543–567. 2023. View Article : Google Scholar : PubMed/NCBI

135 

Xu Y, Hua J, Que H, Zeng T, Li Q, Deng J and Xie J: Identification of PANoptosis-related signature reveals immune infiltration characteristics and immunotherapy responses for renal cell carcinoma. BMC Cancer. 24:2922024. View Article : Google Scholar : PubMed/NCBI

136 

GBD Chronic Kidney Disease Collaboration: Global, regional, and national burden of chronic kidney disease, 1990-2017: A systematic analysis for the global burden of disease study 2017. Lancet. 395:709–733. 2020. View Article : Google Scholar : PubMed/NCBI

137 

Foreman KJ, Marquez N, Dolgert A, Fukutaki K, Fullman N, McGaughey M, Pletcher MA, Smith AE, Tang K, Yuan CW, et al: Forecasting life expectancy, years of life lost, and all-cause and cause-specific mortality for 250 causes of death: Reference and alternative scenarios for 2016-40 for 195 countries and territories. Lancet. 392:2052–2090. 2018. View Article : Google Scholar : PubMed/NCBI

138 

Lin Z, Chen A, Cui H, Shang R, Su T, Li X, Wang K, Yang J, Gao K, Lv J, et al: Renal tubular epithelial cell necroptosis promotes tubulointerstitial fibrosis in patients with chronic kidney disease. FASEB J. 36:e226252022. View Article : Google Scholar : PubMed/NCBI

139 

Zhu Y, Cui H, Xia Y and Gan H: RIPK3-mediated necroptosis and apoptosis contributes to renal tubular cell progressive loss and chronic kidney disease progression in rats. PLoS One. 11:e01567292016. View Article : Google Scholar : PubMed/NCBI

140 

Pang Q, Wang P, Pan Y, Dong X, Zhou T, Song X and Zhang A: Irisin protects against vascular calcification by activating autophagy and inhibiting NLRP3-mediated vascular smooth muscle cell pyroptosis in chronic kidney disease. Cell Death Dis. 13:2832022. View Article : Google Scholar : PubMed/NCBI

141 

Xia Z, Wei Z, Li X, Liu Y, Gu X, Huang S, Zhang X and Wang W: C/EBPα aggravates renal fibrosis in CKD through the NOX4-ROS-apoptosis pathway in tubular epithelial cells. Biochim Biophys Acta Mol Basis Dis. 1870:1670392024. View Article : Google Scholar

142 

Liu X, Liu Z, Wang C, Miao J, Zhou S, Ren Q, Jia N, Zhou L and Liu Y: Kidney tubular epithelial cells control interstitial fibroblast fate by releasing TNFAIP8-encapsulated exosomes. Cell Death Dis. 14:6722023. View Article : Google Scholar : PubMed/NCBI

143 

Huang F, Wang Q, Guo F, Zhao Y, Ji L, An T, Song Y, Liu Y, He Y and Qin G: FoxO1-mediated inhibition of STAT1 alleviates tubulointerstitial fibrosis and tubule apoptosis in diabetic kidney disease. EBioMedicine. 48:491–504. 2019. View Article : Google Scholar : PubMed/NCBI

144 

Cao Y, Chen Z, Hu J, Feng J, Zhu Z, Fan Y, Lin Q and Ding G: Mfn2 regulates high glucose-induced MAMs dysfunction and apoptosis in podocytes via PERK pathway. Front Cell Dev Biol. 9:7692132021. View Article : Google Scholar

145 

Wang Y, Yu L, Li Y, Cha S, Shi L, Wang J, Ge F, Huang C, Huang H, Tu Y, et al: Supplemented gegen qinlian decoction formula attenuates podocyte mitochondrial fission and renal fibrosis in diabetic kidney disease by inhibiting TNF-α-mediated necroptosis, compared with empagliflozin. J Ethnopharmacol. 334:1185722024. View Article : Google Scholar

146 

Yu Q, Chen Y, Zhao Y, Huang S, Xin X, Jiang L, Wang H, Wu W, Qu L, Xiang C, et al: Nephropathy is aggravated by fatty acids in diabetic kidney disease through tubular epithelial cell necroptosis and is alleviated by an RIPK-1 inhibitor. Kidney Dis (Basel). 9:408–423. 2023. View Article : Google Scholar : PubMed/NCBI

147 

Shi Y, Chen X, Huang C and Pollock C: RIPK3: A new player in renal fibrosis. Front Cell Dev Biol. 8:5022020. View Article : Google Scholar : PubMed/NCBI

148 

Hu B, Ma K, Wang W, Han Z, Chi M, Nasser MI and Liu C: Research progress of pyroptosis in renal diseases. Curr Med Chem. 31:6656–6671. 2024. View Article : Google Scholar

149 

Liu P, Zhang Z and Li Y: Relevance of the pyroptosis-related inflammasome pathway in the pathogenesis of diabetic kidney disease. Front Immunol. 12:6034162021. View Article : Google Scholar : PubMed/NCBI

150 

Zhang WT, Ge HW, Wei Y, Gao JL, Tian F and Zhou EC: Molecular characterization of PANoptosis-related genes in chronic kidney disease. PLoS One. 19:e03126962024. View Article : Google Scholar : PubMed/NCBI

151 

Huang SC, Zhang L, Sung YS, Chen CL, Krausz T, Dickson BC, Kao YC, Agaram NP, Fletcher CD and Antonescu CR: Frequent FOS gene rearrangements in epithelioid hemangioma: A molecular study of 58 cases with morphologic reappraisal. Am J Surg Pathol. 39:1313–1321. 2015. View Article : Google Scholar : PubMed/NCBI

152 

Durchdewald M, Angel P and Hess J: The transcription factor Fos: A Janus-type regulator in health and disease. Histol Histopathol. 24:1451–1461. 2009.PubMed/NCBI

153 

Huang YS, Lo CH, Tsai PH, Hou YC, Chang YT, Guo CY, Hsieh HY, Lu KC, Shih HM and Wu CC: Downregulation of AANAT by c-Fos in tubular epithelial cells with membranous nephropathy. Biochem Biophys Res Commun. 584:32–38. 2021. View Article : Google Scholar : PubMed/NCBI

154 

Wang F, Wang S, Wang J, Huang K, Chen G, Peng Y, Liu C and Tao Y: Pharmacological mechanisms of Fuzheng Huayu formula for Aristolochic acid I-induced kidney fibrosis through network pharmacology. Front Pharmacol. 13:10568652022. View Article : Google Scholar : PubMed/NCBI

155 

Yang G, Wang M, Qahar M, He J, Lai Z, Li S, He D, Yan X, Xiong Z, Xiong Z and Le TH: Post-burns persistent inflammation leads to kidney PANoptosis with caspases pathway activation. Heliyon. 11:e414852024. View Article : Google Scholar

156 

Lv Z, Hu J, Su H, Yu Q, Lang Y, Yang M, Fan X, Liu Y, Liu B, Zhao Y, et al: TRAIL induces podocyte PANoptosis via death receptor 5 in diabetic kidney disease. Kidney Int. 107:317–331. 2025. View Article : Google Scholar

157 

Zhang HR, Li YP, Shi ZJ, Liang QQ, Chen SY, You YP, Yuan T, Xu R, Xu LH, Ouyang DY, et al: Triptolide induces PANoptosis in macrophages and causes organ injury in mice. Apoptosis. 28:1646–1665. 2023. View Article : Google Scholar : PubMed/NCBI

158 

Xu C, Wang Q, Du C, Chen L, Zhou Z, Zhang Z, Cai N, Li J, Huang C and Ma T: Histone deacetylase-mediated silencing of PSTPIP2 expression contributes to aristolochic acid nephropathy-induced PANoptosis. Br J Pharmacol. 181:1452–1473. 2024. View Article : Google Scholar

159 

Yuan F, Cai J, Wu J, Tang Y, Zhao K, Liang F, Li F, Yang X, He Z, Billiar TR, et al: Z-DNA binding protein 1 promotes heatstroke-induced cell death. Science. 376:609–615. 2022. View Article : Google Scholar : PubMed/NCBI

160 

Sanz AB, Sanchez-Niño MD, Ramos AM and Ortiz A: Regulated cell death pathways in kidney disease. Nat Rev Nephrol. 19:281–299. 2023. View Article : Google Scholar : PubMed/NCBI

161 

Ruiz-Ortega M, Rayego-Mateos S, Lamas S, Ortiz A and Rodrigues-Diez RR: Targeting the progression of chronic kidney disease. Nat Rev Nephrol. 16:269–288. 2020. View Article : Google Scholar : PubMed/NCBI

162 

Weisel K, Berger S, Thorn K, Taylor PC, Peterfy C, Siddall H, Tompson D, Wang S, Quattrocchi E, Burriss SW, et al: A randomized, placebo-controlled experimental medicine study of RIPK1 inhibitor GSK2982772 in patients with moderate to severe rheumatoid arthritis. Arthritis Res Ther. 23:852021. View Article : Google Scholar : PubMed/NCBI

163 

Weisel K, Scott N, Berger S, Wang S, Brown K, Powell M, Broer M, Watts C, Tompson DJ, Burriss SW, et al: A randomised, placebo-controlled study of RIPK1 inhibitor GSK2982772 in patients with active ulcerative colitis. BMJ Open Gastroenterol. 8:e0006802021. View Article : Google Scholar : PubMed/NCBI

164 

Shiffman M, Freilich B, Vuppalanchi R, Watt K, Chan JL, Spada A, Hagerty DT and Schiff E: Randomised clinical trial: emricasan versus placebo significantly decreases ALT and caspase 3/7 activation in subjects with non-alcoholic fatty liver disease. Aliment Pharmacol Ther. 49:64–73. 2019. View Article : Google Scholar

165 

Ludbrook VJ, Budd DC, Thorn K, Tompson D, Votta BJ, Walker L, Lee A, Chen X, Peppercorn A and Loo WJ: Inhibition of receptor-interacting protein kinase 1 in chronic plaque psoriasis: A multicenter, randomized, double-blind, placebo-controlled study. Dermatol Ther (Heidelb). 14:489–504. 2024. View Article : Google Scholar : PubMed/NCBI

166 

Weisel K, Berger S, Papp K, Maari C, Krueger JG, Scott N, Tompson D, Wang S, Simeoni M, Bertin J and Peter Tak P: Response to inhibition of receptor-interacting protein kinase 1 (RIPK1) in active plaque psoriasis: A randomized placebo-controlled study. Clin Pharmacol Ther. 108:808–816. 2020. View Article : Google Scholar : PubMed/NCBI

167 

Karki R, Sundaram B, Sharma BR, Lee S, Malireddi RKS, Nguyen LN, Christgen S, Zheng M, Wang Y, Samir P, et al: ADAR1 restricts ZBP1-mediated immune response and PANoptosis to promote tumorigenesis. Cell Rep. 37:1098582021. View Article : Google Scholar : PubMed/NCBI

168 

Oh S, Lee J, Oh J, Yu G, Ryu H, Kim D and Lee S: Integrated NLRP3, AIM2, NLRC4, Pyrin inflammasome activation and assembly drive PANoptosis. Cell Mol Immunol. 20:1513–1526. 2023. View Article : Google Scholar : PubMed/NCBI

169 

Huang R, Hu Y, Wang YF, Zhang S, Wang ZG, Pang DW and Liu SL: Targeted degradation of ZBP1 with covalent PROTACs for anti-inflammatory treatment of infections. Angew Chem Int Ed Engl. 64:e2024235242025. View Article : Google Scholar : PubMed/NCBI

170 

Zhang T, Yin C, Fedorov A, Qiao L, Bao H, Beknazarov N, Wang S, Gautam A, Williams RM, Crawford JC, et al: ADAR1 masks the cancer immunotherapeutic promise of ZBP1-driven necroptosis. Nature. 606:594–602. 2022. View Article : Google Scholar : PubMed/NCBI

171 

Yang W, Tao K, Wang Y, Huang Y, Duan C, Wang T, Li C, Zhang P, Yin Y, Gao J and Li R: Necrosulfonamide ameliorates intestinal inflammation via inhibiting GSDMD-medicated pyroptosis and MLKL-mediated necroptosis. Biochem Pharmacol. 206:1153382022. View Article : Google Scholar : PubMed/NCBI

172 

Green JP, El-Sharkawy LY, Roth S, Zhu J, Cao J, Leach AG, Liesz A, Freeman S and Brough D: Discovery of an inhibitor of DNA-driven inflammation that preferentially targets the AIM2 inflammasome. iScience. 26:1067582023. View Article : Google Scholar : PubMed/NCBI

173 

Lin Q, Li S, Jiang N, Jin H, Shao X, Zhu X, Wu J, Zhang M, Zhang Z, Shen J, et al: Inhibiting NLRP3 inflammasome attenuates apoptosis in contrast-induced acute kidney injury through the upregulation of HIF1A and BNIP3-mediated mitophagy. Autophagy. 17:2975–2990. 2021. View Article : Google Scholar :

174 

Li C, Yang H, Wu Y, Zhou M, Luo H, Yuan P and Shen F: Carnosol alleviates cisplatin-induced acute kidney injury by regulating apoptosis and pyroptosis. Cell Biol Int. 49:101–117. 2025. View Article : Google Scholar

175 

Chen L, Fang H, Li X, Yu P, Guan Y, Xiao C, Deng Z, Hei Z, Chen C and Luo C: Connexin32 gap junction channels deliver miR155-3p to mediate pyroptosis in renal ischemia-reperfusion injury. Cell Commun Signal. 22:1212024. View Article : Google Scholar : PubMed/NCBI

176 

Li J, Lin Q, Shao X, Li S, Zhu X, Wu J, Mou S, Gu L, Wang Q, Zhang M, et al: HIF1α-BNIP3-mediated mitophagy protects against renal fibrosis by decreasing ROS and inhibiting activation of the NLRP3 inflammasome. Cell Death Dis. 14:2002023. View Article : Google Scholar

177 

Ji Y, Hua H, Jia Z, Zhang A and Ding G: Therapy targeted to the NLRP3 inflammasome in chronic kidney disease. Kidney Dis (Basel). 10:369–383. 2024. View Article : Google Scholar : PubMed/NCBI

178 

Østergaard JA, Jha JC, Sharma A, Dai A, Choi JSY, de Haan JB, Cooper ME and Jandeleit-Dahm K: Adverse renal effects of NLRP3 inflammasome inhibition by MCC950 in an interventional model of diabetic kidney disease. Clin Sci (Lond). 136:167–180. 2022. View Article : Google Scholar : PubMed/NCBI

179 

Wu M, Yang Z, Zhang C and Shi Y, Han W, Song S, Mu L, Du C and Shi Y: Inhibition of NLRP3 inflammasome ameliorates podocyte damage by suppressing lipid accumulation in diabetic nephropathy. Metabolism. 118:1547482021. View Article : Google Scholar : PubMed/NCBI

180 

Chen YM, Tang BX, Chen WY and Zhao MS: Ursolic acid inhibits the invasiveness of A498 cells via NLRP3 inflammasome activation. Oncol Lett. 20:1702020. View Article : Google Scholar : PubMed/NCBI

181 

Klück V, Jansen TLTA, Janssen M, Comarniceanu A, Efdé M, Tengesdal IW, Schraa K, Cleophas MCP, Scribner CL, Skouras DB, et al: Dapansutrile, an oral selective NLRP3 inflammasome inhibitor, for treatment of gout flares: An open-label, dose-adaptive, proof-of-concept, phase 2a trial. Lancet Rheumatol. 2:e270–e280. 2020. View Article : Google Scholar : PubMed/NCBI

182 

Yin P, Jia J, Li J, Song Y, Zhang Y and Chen F: ABT-737, a Bcl-2 selective inhibitor, and chloroquine synergistically kill renal cancer cells. Oncol Res. 24:65–72. 2016. View Article : Google Scholar : PubMed/NCBI

183 

Sun H, Wang W, Che Y and Jiang X: Fungal secondary metabolites rasfonin induces autophagy, apoptosis and necroptosis in renal cancer cell line. Mycology. 7:81–87. 2016. View Article : Google Scholar : PubMed/NCBI

184 

Ding Z, Zhao J, Wang X, Li W, Chen C, Yong C, Zhu Y, Tian F, Liu L, Yu M, et al: Total extract of Abelmoschus manihot L. alleviates uric acid-induced renal tubular epithelial injury via inhibition of caspase-8/caspase-3/NLRP3/GSDME signaling. Front Pharmacol. 13:9079802022. View Article : Google Scholar : PubMed/NCBI

185 

Li X, Hu D, Li Y, Luo Y, Liang B, Yu K, Xiong W and Zuo D: Overexpression of TP53INP2 promotes apoptosis in clear cell renal cell cancer via caspase-8/TRAF6 signaling pathway. J Immunol Res. 2022:12604232022.PubMed/NCBI

186 

Gautam A, Boyd DF, Nikhar S, Zhang T, Siokas I, Van de Velde LA, Gaevert J, Meliopoulos V, Thapa B, Rodriguez DA, et al: Necroptosis blockade prevents lung injury in severe influenza. Nature. 628:835–843. 2024. View Article : Google Scholar : PubMed/NCBI

187 

Abou Taha MA, Ali FEM, Saleh IG and Akool ES: Sorafenib and edaravone protect against renal fibrosis induced by unilateral ureteral obstruction via inhibition of oxidative stress, inflammation, and RIPK-3/MLKL pathway. Naunyn Schmiedebergs Arch Pharmacol. 397:8961–8977. 2024. View Article : Google Scholar : PubMed/NCBI

188 

Hou Y, Feng Q, Wei C, Cao F, Liu D, Pan S, Shi Y, Liu Z and Liu F: Emerging role of PANoptosis in kidney diseases: Molecular mechanisms and therapeutic opportunities. Apoptosis. 30:579–596. 2025. View Article : Google Scholar : PubMed/NCBI

Related Articles

  • Abstract
  • View
  • Download
  • Twitter
Copy and paste a formatted citation
Spandidos Publications style
Jiang L, Zhao L, Liu Y, Fu Z, Wu M, Mou Z and Li M: PANoptosis: A novel therapeutic target in kidney disease (Review). Int J Mol Med 56: 207, 2025.
APA
Jiang, L., Zhao, L., Liu, Y., Fu, Z., Wu, M., Mou, Z., & Li, M. (2025). PANoptosis: A novel therapeutic target in kidney disease (Review). International Journal of Molecular Medicine, 56, 207. https://doi.org/10.3892/ijmm.2025.5648
MLA
Jiang, L., Zhao, L., Liu, Y., Fu, Z., Wu, M., Mou, Z., Li, M."PANoptosis: A novel therapeutic target in kidney disease (Review)". International Journal of Molecular Medicine 56.6 (2025): 207.
Chicago
Jiang, L., Zhao, L., Liu, Y., Fu, Z., Wu, M., Mou, Z., Li, M."PANoptosis: A novel therapeutic target in kidney disease (Review)". International Journal of Molecular Medicine 56, no. 6 (2025): 207. https://doi.org/10.3892/ijmm.2025.5648
Copy and paste a formatted citation
x
Spandidos Publications style
Jiang L, Zhao L, Liu Y, Fu Z, Wu M, Mou Z and Li M: PANoptosis: A novel therapeutic target in kidney disease (Review). Int J Mol Med 56: 207, 2025.
APA
Jiang, L., Zhao, L., Liu, Y., Fu, Z., Wu, M., Mou, Z., & Li, M. (2025). PANoptosis: A novel therapeutic target in kidney disease (Review). International Journal of Molecular Medicine, 56, 207. https://doi.org/10.3892/ijmm.2025.5648
MLA
Jiang, L., Zhao, L., Liu, Y., Fu, Z., Wu, M., Mou, Z., Li, M."PANoptosis: A novel therapeutic target in kidney disease (Review)". International Journal of Molecular Medicine 56.6 (2025): 207.
Chicago
Jiang, L., Zhao, L., Liu, Y., Fu, Z., Wu, M., Mou, Z., Li, M."PANoptosis: A novel therapeutic target in kidney disease (Review)". International Journal of Molecular Medicine 56, no. 6 (2025): 207. https://doi.org/10.3892/ijmm.2025.5648
Follow us
  • Twitter
  • LinkedIn
  • Facebook
About
  • Spandidos Publications
  • Careers
  • Cookie Policy
  • Privacy Policy
How can we help?
  • Help
  • Live Chat
  • Contact
  • Email to our Support Team