1
|
Yao M, Miao L, Wang X, Han Y. Targeting programmed cell death pathways in gastric cancer: a focus on pyroptosis, apoptosis, necroptosis and PANoptosis. Gene 2025; 960:149546. [PMID: 40334955 DOI: 10.1016/j.gene.2025.149546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 04/05/2025] [Accepted: 05/02/2025] [Indexed: 05/09/2025]
Abstract
Gastric cancer (GC) is recognized as one of the most prevalent and serious malignancies, distinguished by its high incidence and fatality rates. Given the considerable mortality rate associated with GC, it is imperative to clarify the related pathways of GC development and further identify feasible targets for rational targeted therapy. Accumulating evidence reveals that programmed cell death (PCD) is a crucial element in both the progression and treatment of cancer. Pyroptosis, apoptosis, and necroptosis are three well-studied types of PCD, and a link between them and GC has been established in recent studies. PANoptosis, a comparatively novel type of PCD, shares key traits with pyroptosis, apoptosis, and necroptosis, yet cannot be entirely illustrated by any single model. PANoptosis has been discovered to exert an impact on multiple diseases, including cancer, infections, and inflammatory conditions, consequently offering novel perceptions into the progression and treatment of GC. This review seeks to encapsulate the emerging roles and therapeutic potential of pyroptosis, apoptosis, necroptosis, and PANoptosis in GC, laying the groundwork for the advancement of innovative treatment methods that target important signaling pathways connected with these four forms of PCD.
Collapse
Affiliation(s)
- Minghui Yao
- Department of Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi 830017, China
| | - Liying Miao
- Department of Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi 830017, China
| | - Xin Wang
- Department of Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi 830017, China
| | - Yangyang Han
- Department of Biology, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi 830017, China; Xinjiang Key Laboratory of Molecular Biology for Endemic Diseases, Xinjiang Medical University, Urumqi 830017, China.
| |
Collapse
|
2
|
Song R, Yin S, Wu J, Yan J. Neuronal regulated cell death in aging-related neurodegenerative diseases: key pathways and therapeutic potentials. Neural Regen Res 2025; 20:2245-2263. [PMID: 39104166 PMCID: PMC11759035 DOI: 10.4103/nrr.nrr-d-24-00025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 05/13/2024] [Accepted: 06/18/2024] [Indexed: 08/07/2024] Open
Abstract
Regulated cell death (such as apoptosis, necroptosis, pyroptosis, autophagy, cuproptosis, ferroptosis, disulfidptosis) involves complex signaling pathways and molecular effectors, and has been proven to be an important regulatory mechanism for regulating neuronal aging and death. However, excessive activation of regulated cell death may lead to the progression of aging-related diseases. This review summarizes recent advances in the understanding of seven forms of regulated cell death in age-related diseases. Notably, the newly identified ferroptosis and cuproptosis have been implicated in the risk of cognitive impairment and neurodegenerative diseases. These forms of cell death exacerbate disease progression by promoting inflammation, oxidative stress, and pathological protein aggregation. The review also provides an overview of key signaling pathways and crosstalk mechanisms among these regulated cell death forms, with a focus on ferroptosis, cuproptosis, and disulfidptosis. For instance, FDX1 directly induces cuproptosis by regulating copper ion valency and dihydrolipoamide S-acetyltransferase aggregation, while copper mediates glutathione peroxidase 4 degradation, enhancing ferroptosis sensitivity. Additionally, inhibiting the Xc- transport system to prevent ferroptosis can increase disulfide formation and shift the NADP + /NADPH ratio, transitioning ferroptosis to disulfidptosis. These insights help to uncover the potential connections among these novel regulated cell death forms and differentiate them from traditional regulated cell death mechanisms. In conclusion, identifying key targets and their crosstalk points among various regulated cell death pathways may aid in developing specific biomarkers to reverse the aging clock and treat age-related neurodegenerative conditions.
Collapse
Affiliation(s)
- Run Song
- Department of Neurology, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
- Neuromolecular Biology Laboratory, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
| | - Shiyi Yin
- Department of Neurology, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
- Neuromolecular Biology Laboratory, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
| | - Jiannan Wu
- Neuromolecular Biology Laboratory, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
| | - Junqiang Yan
- Department of Neurology, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
- Neuromolecular Biology Laboratory, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, Henan Province, China
| |
Collapse
|
3
|
Qi H, Zhang X, Zhang Z, Gao Y, Tian D, Zhao G, Xie Z, Zeng J, Zhang L, Zeng N, Yang R. The extract of chrysanthemum flos mitigates post-stroke sarcopenia by inhibiting PANoptosis and restoring muscle homeostasis. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156784. [PMID: 40311590 DOI: 10.1016/j.phymed.2025.156784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 04/09/2025] [Accepted: 04/17/2025] [Indexed: 05/03/2025]
Abstract
BACKGROUND Sarcopenia and muscle weakness are prevalent complications of ischemic stroke (IS), with limited pharmacological options. This study identifies high-dose extracts of Chrysanthemum Flos (ECF) as a potential therapy for post-stroke muscle dysfunction by targeting PANoptosis-a pro-inflammatory programmed cell death pathway. Through its anti-inflammatory and antioxidant properties, ECF attenuates muscle atrophy and enhances functional recovery, offering novel insights into ISS treatment. PURPOSE To evaluate the therapeutic efficacy of high-dose ECF in ischemic stroke-induced sarcopenia (ISS) and elucidate its regulatory role in PANoptosis-mediated muscle degeneration and protein homeostasis. METHODS Preparation of a rat middle cerebral artery occlusion (MCAO) model using intravascular wire thrombus blockade. Cerebral injury was assessed using laser speckle contrast imaging, triphenyltetrazolium chloride (TTC) staining, and Zea-Longa neurological scoring. ECF's effects on muscle function were evaluated through gait analysis, muscle morphology (length and weight), grip strength, electromyography, and H&E staining. RNA sequencing was conducted to elucidate transcriptomic alterations and enriched pathways associated with ECF in ISS. PANoptosis-mediated myofiber and L6 cell damage was analyzed by flow cytometry (FC), immunofluorescence (IF), immunohistochemistry (IHC), and western blotting (WB). ECF composition and quality were validated using liquid chromatography-mass spectrometry (LC-MS). RESULTS ISS rats showed 83 % reductions in endurance, grip strength, and EMG signals compared to sham (p < 0.01), which improved to 70 % of normal after ECF treatment. ECF significantly increased muscle fiber area, alleviated mitochondrial damage, and improved sarcomere structure (p < 0.001). RNA-seq identified TNF signaling and PANoptosis (apoptosis, pyroptosis, necroptosis) as key drivers of ISS-induced muscle injury. The TNF-targeted inhibitor R7050 further confirmed TNF-α as a critical activator of Z-DNA binding protein 1 (ZBP1). ECF treatment significantly reduced tissue inflammation (p < 0.01) and inhibited ZBP1 expression (p < 0.01). Following ISS, key PANoptosis-related proteins, including ZBP1, Gasdermin D N-terminal fragment (GSDMD-N), Cleaved-Caspase3, Caspase6, Caspase8, phosphorylated mixed-lineage kinase domain-like (p-MLKL), Phosphorylated Receptor-Interacting Protein Kinase 1 (p-RIPK1), Phosphorylated Receptor-Interacting Protein Kinase 3 (p-RIPK3), and NOD-like receptor family pyrin domain containing 3 (NLRP3), were significantly upregulated (p < 0.05), while ECF-H treatment significantly suppressed their expression (p < 0.05, p < 0.01). Additionally, ECF significantly promoted the expression of muscle protein synthesis factors (myogenic differentiation 1 (MyoD1) and recombinant myosin heavy chain 1 (MYH), p < 0.01) and inhibited protein degradation factors (muscle RING-finger protein-1 (MuRF1) and muscle atrophy F-Box protein (MAFbx), p < 0.01), thus maintaining muscle protein homeostasis. The results from PCR, WB, IHC, IF, and FC experiments were consistent with RNA-seq findings. CONCLUSIONS ECF ameliorates ISS in MCAO rats by inhibiting muscle PANoptosis, which simultaneously reduces protein degradation and enhances protein synthesis.
Collapse
Affiliation(s)
- Hu Qi
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Xiongwei Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Zeyang Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Yuanlin Gao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Dan Tian
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Ge Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Zhiqiang Xie
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Jiuseng Zeng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Lihong Zhang
- Department of Otorhinolaryngology, Chengdu Xinjin District Hospital of Traditional Chinese Medicine, Chengdu, Sichuan Province, China
| | - Nan Zeng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China.
| | - Ruocong Yang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China; School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan Province, China.
| |
Collapse
|
4
|
Ren H, Zhao C, Zhou L, Ke Q, Chen Y, Chen Z, He J, Chen D, He X, Quan M, Liu L, Li R, Pan H. Licochalcone B attenuates pulmonary fibrosis via inhibiting ZBP1-dependent PANoptosis. JOURNAL OF ETHNOPHARMACOLOGY 2025; 349:119940. [PMID: 40350052 DOI: 10.1016/j.jep.2025.119940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 05/01/2025] [Accepted: 05/07/2025] [Indexed: 05/14/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Pulmonary fibrosis (PF) is a chronic, progressive, and frequently fatal interstitial lung disease. Glycyrrhiza uralensis Fisch, a traditional Chinese medicine (TCM) herb, has long been used for respiratory disorders due to its anti-inflammatory and expectorant properties. Licochalcone B (LCB), a chalcone derivative isolated from Glycyrrhiza uralensis Fisch, shows therapeutic potential in airway inflammation and alveolar injury, making it a promising candidate for fibrotic lung diseases. AIM OF THE STUDY This study evaluates the anti-fibrotic efficacy of LCB against PF progression and elucidates its potential molecular mechanisms. MATERIALS AND METHODS PF was induced in mice by intratracheal administration of bleomycin (BLM, 1.25 mg/kg). After disease model induction, the mice were treated with LCB (3.13 or 6.25 mg/kg per day) or pirfenidone (PFD, 300 mg/kg per day) for 14 days. Histopathological changes, collagen deposition, fibrosis-related factor levels, and PANoptotic marker protein in lung tissue were evaluated. Two TGF-β1-induced PF cell models (NIH-3T3 and BEAS-2B) were utilized to simulate fibroblast activation and epithelial-mesenchymal transition processes. The effects of LCB intervention on cell proliferation, migration, and the expression of vimentin, fibronectin (FN), and type I collagen (COL1A1) were assessed through wound healing assays, colony formation assays, Western blotting (WB), and immunocytochemistry. RESULTS LCB significantly alleviates BLM-induced pulmonary inflammation and fibrosis, reduces collagen deposition in lung tissues of fibrotic mice, and downregulates FN expression while upregulating E-cadherin (E-Cad) levels. Immunohistochemical analysis revealed that LCB downregulates the expression of ZBP1 and apoptotic marker proteins in the lung tissues of PF mice. Additionally, LCB inhibits TGF-β1-induced abnormal migration of BEAS-2B cells and aberrant proliferation of NIH/3T3 cells while suppressing the expression of fibrosis-related factors, including COL1A1, FN, and α-smooth muscle actin (α-SMA). Our findings demonstrate that ZBP1 overexpression in epithelial cells attenuates the anti-fibrotic efficacy of LCB through the activation of PANoptosis and identify the inhibition of IRF1 binding to the ZBP1 promoter as a pivotal mechanism underlying the therapeutic potential of LCB in PF. CONCLUSION This study found that LCB exerts pleiotropic antifibrotic effects by targeting ZBP1-mediated PANoptosis, thereby identifying ZBP1 as a critical therapeutic target for PF and highlighting LCB's potential in anti-fibrotic therapy.
Collapse
Affiliation(s)
- Hong Ren
- State Key Laboratory of Traditional Chinese Medicine Syndrome/ The Second Clinical College, Guangzhou University of Chinese Medicine, Guangdong Province, 510405, China
| | - Caiping Zhao
- State Key Laboratory of Traditional Chinese Medicine Syndrome/ The Second Clinical College, Guangzhou University of Chinese Medicine, Guangdong Province, 510405, China; Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510120, China
| | - Lvzhou Zhou
- State Key Laboratory of Traditional Chinese Medicine Syndrome/ The Second Clinical College, Guangzhou University of Chinese Medicine, Guangdong Province, 510405, China
| | - Qingming Ke
- State Key Laboratory of Traditional Chinese Medicine Syndrome/ The Second Clinical College, Guangzhou University of Chinese Medicine, Guangdong Province, 510405, China
| | - Yulian Chen
- State Key Laboratory of Traditional Chinese Medicine Syndrome/ The Second Clinical College, Guangzhou University of Chinese Medicine, Guangdong Province, 510405, China
| | - Zhengmin Chen
- State Key Laboratory of Traditional Chinese Medicine Syndrome/ The Second Clinical College, Guangzhou University of Chinese Medicine, Guangdong Province, 510405, China
| | - Jiayan He
- State Key Laboratory of Traditional Chinese Medicine Syndrome/ The Second Clinical College, Guangzhou University of Chinese Medicine, Guangdong Province, 510405, China
| | - Danli Chen
- State Key Laboratory of Traditional Chinese Medicine Syndrome/ The Second Clinical College, Guangzhou University of Chinese Medicine, Guangdong Province, 510405, China
| | - Xizi He
- State Key Laboratory of Traditional Chinese Medicine Syndrome/ The Second Clinical College, Guangzhou University of Chinese Medicine, Guangdong Province, 510405, China
| | - Minqi Quan
- State Key Laboratory of Traditional Chinese Medicine Syndrome/ The Second Clinical College, Guangzhou University of Chinese Medicine, Guangdong Province, 510405, China
| | - Liang Liu
- State Key Laboratory of Traditional Chinese Medicine Syndrome/ The Second Clinical College, Guangzhou University of Chinese Medicine, Guangdong Province, 510405, China; Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510120, China; Chinese Medicine Guangdong Laboratory/ Hengqin Laboratory, Hengqin, 519031, Guangdong, China.
| | - Runze Li
- State Key Laboratory of Traditional Chinese Medicine Syndrome/ The Second Clinical College, Guangzhou University of Chinese Medicine, Guangdong Province, 510405, China; Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510120, China; Chinese Medicine Guangdong Laboratory/ Hengqin Laboratory, Hengqin, 519031, Guangdong, China.
| | - Hudan Pan
- State Key Laboratory of Traditional Chinese Medicine Syndrome/ The Second Clinical College, Guangzhou University of Chinese Medicine, Guangdong Province, 510405, China; Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, 510120, China; Chinese Medicine Guangdong Laboratory/ Hengqin Laboratory, Hengqin, 519031, Guangdong, China.
| |
Collapse
|
5
|
Zhang YT, Li HH, Teng F. Inhibition of Piezo1 ameliorates septic cardiomyopathy by blocking calcium-dependent PANoptosis. Eur J Pharmacol 2025; 996:177438. [PMID: 40020983 DOI: 10.1016/j.ejphar.2025.177438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 02/14/2025] [Accepted: 02/26/2025] [Indexed: 03/03/2025]
Abstract
Sepsis-induced cardiomyopathy (SIC) represents a severe and often fatal complication of sepsis, characterized by significant mortality. Despite extensive research, the underlying mechanisms remain incompletely understood. Recent studies have highlighted PANoptosis, an emerging form of programmed cell death, as a critical factor in inflammatory diseases. Piezo1, a mechanosensitive ion channel, has been implicated in various pathological conditions; however, its role in SIC and its involvement in PANoptosis require further investigation. In this study, the role of Piezo1 in SIC and calcium-dependent PANoptosis were investigated. SIC was induced in mice via cecal ligation and puncture (CLP), and the effects of Piezo1 inhibition on cardiac function, histological changes, mitochondrial function, and PANoptosis were assessed. Our results show that sepsis upregulates Piezo1 expression in cardiomyocytes through TLR4-NF-κB signaling. Pharmacological blockade of Piezo1 with its inhibitor GsMTx4 attenuated CLP-induced cardiac injury, histological damage, and mitochondrial dysfunction. Importantly, Piezo1 inhibition also significantly suppressed PANoptosis in septic hearts. In vitro experiments with Piezo1 siRNA, GsMTx4 and the calcium chelator BAPTA confirmed that inhibition of Piezo1 attenuates LPS-induced PANoptosis by limiting calcium release in cardiomyocytes after LPS treatment, linking Piezo1 to the regulation of these key events. Collectively, these findings reveal Piezo1 as a novel mechanosensor for sepsis and reveal a previously unrecognized role of Piezo1 in the activation of calcium-mediated PANoptosis in SIC. Given the ability of Piezo1 inhibition to mitigate key pathological features of SIC, targeting Piezo1 represents a promising therapeutic strategy for improving the outcomes of sepsis-related cardiac dysfunction.
Collapse
Affiliation(s)
- Yan-Ting Zhang
- Medical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Hui-Hua Li
- Medical Research Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China.
| | - Fei Teng
- Emergency Medicine Center, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China.
| |
Collapse
|
6
|
Cui Y, Lin H, Ma J, Zhao Y, Li J, Wang Y, Zhuang J, Yang Y. Ischemia-reperfusion injury induces ZBP1-dependent PANoptosis in endothelial cells. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167782. [PMID: 40057207 DOI: 10.1016/j.bbadis.2025.167782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 02/02/2025] [Accepted: 03/04/2025] [Indexed: 04/15/2025]
Abstract
Endothelial cells play a critical role in the pathophysiology of ischemia-reperfusion injury (IRI). Although previous studies have shown that IRI can activate PANoptosis, the underlying mechanisms remain unclear. Our research investigates how IRI induces PANoptosis in endothelial cells, aiming to identify protective strategies to safeguard these cells from PANoptosis triggered by IRI. We established an in vitro endothelial cell hypoxia/reoxygenation (H/R) treatment model and an in vivo SD rat free flap IRI model. A series of assays, including PI/Hoechst staining, Western blotting, and immunohistochemistry, were conducted to assess PANoptosis-like cell death in endothelial cells. Cell transfection with ZBP1 siRNA and immunoprecipitation were used to explore the involved signaling pathways. Our results showed activation of PANoptosis-like cell death and upregulation of ZBP1 expression following IRI. After knocking down ZBP1 expression, a significant alteration in PANoptosis-like cell death and the assembly of the ZBP1-PANoptosome in endothelial cells was observed, confirming the occurrence of PANoptosis. In conclusion, our research confirms that IRI induces PANoptosome formation, promoting ZBP1-dependent PANoptosis in endothelial cells.
Collapse
Affiliation(s)
- Yue Cui
- Department of Aesthetic Plastic Surgery and Laser Medicine, Beijing Anzhen Hospital Affiliated to Capital Medical University, Beijing 100029, China
| | - Huang Lin
- Department of Aesthetic Plastic Surgery and Laser Medicine, Beijing Anzhen Hospital Affiliated to Capital Medical University, Beijing 100029, China.
| | - Jiaxing Ma
- Department of Aesthetic Plastic Surgery and Laser Medicine, Beijing Anzhen Hospital Affiliated to Capital Medical University, Beijing 100029, China
| | - Yinhua Zhao
- Department of Aesthetic Plastic Surgery and Laser Medicine, Beijing Anzhen Hospital Affiliated to Capital Medical University, Beijing 100029, China
| | - Jiaxi Li
- Department of Aesthetic Plastic Surgery and Laser Medicine, Beijing Anzhen Hospital Affiliated to Capital Medical University, Beijing 100029, China
| | - Yang Wang
- Department of Aesthetic Plastic Surgery and Laser Medicine, Beijing Anzhen Hospital Affiliated to Capital Medical University, Beijing 100029, China
| | - Jingwei Zhuang
- Department of Aesthetic Plastic Surgery and Laser Medicine, Beijing Anzhen Hospital Affiliated to Capital Medical University, Beijing 100029, China
| | - Yu Yang
- Department of Plastic Surgery, Fujian Provincial Hospital, Provincial Clinical College of Fujian Medical University, Fuzhou 350001, China
| |
Collapse
|
7
|
Hou K, Pan W, Liu L, Yu Q, Ou J, Li Y, Yang X, Lin Z, Yuan JH, Fang M. Molecular mechanism of PANoptosis and programmed cell death in neurological diseases. Neurobiol Dis 2025; 209:106907. [PMID: 40204169 DOI: 10.1016/j.nbd.2025.106907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/03/2025] [Accepted: 04/06/2025] [Indexed: 04/11/2025] Open
Abstract
PANoptosis represents a highly coordinated inflammatory programmed cell death governed by the assembly and activation of PANoptosome, which strategically integrate core molecular elements from pyroptosis, apoptosis, and necroptosis. The triple-component cell death pathways set themselves apart from alternative regulated cell death mechanisms through their unique capacity to concurrently integrate and process molecular signals derived from multiple death-signaling modalities, thereby coordinating a multifaceted cellular defense system against diverse pathological insults. Pathogen-associated molecular patterns synergistically interact with cytokine storms, and oncogenic stress to active PANoptosis, establishing this programmed cell death pathway as a critical nexus in inflammatory pathogenesis and tumor immunomodulation. This molecular crosstalk highlights PANoptosis as a promising therapeutic target for managing immune-related disorders and malignant transformation. Emerging evidence links PANoptosis to neuroinflammatory disorders through dysregulated crosstalk between programmed death pathways (apoptosis, necroptosis, pyroptosis) and accidental necrosis, driving neuronal loss and neural damage. Single-cell transcriptomics reveals spatially resolved PANoptosis signatures in Alzheimer's hippocampal microenvironments and multiple sclerosis demyelinating plaques, with distinct molecular clusters correlating to quantifiable neuroinflammatory metrics. Emerging PANoptosis-targeted therapies show preclinical promise in alleviating neurovascular dysfunction while preserving physiological microglial surveillance functions. Accumulating evidence linking dysregulated cell death pathways (particularly PANoptosis) to neurological disorders underscores the urgency of deciphering its molecular mechanisms and developing precision modulators as next-generation therapies. This review systematically deciphers PANoptosome assembly mechanisms and associated cell death cascades, evaluates their pathological roles in neurological disorders through multiscale regulatory networks, and proposes PANoptosis-targeted therapeutic frameworks to advance precision neurology.
Collapse
Affiliation(s)
- Ketian Hou
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wenhan Pan
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lianhui Liu
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qianqian Yu
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jiahao Ou
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yueqi Li
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xi Yang
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhenlang Lin
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, China; Key Laboratory of Perinatal Medicine of Wenzhou, Wenzhou, Zhejiang, China.
| | - Jun Hui Yuan
- Department of Neonatology, Wenling Maternal and Child Health Care Hospital, Wenling, Zhejiang 317500, China.
| | - Mingchu Fang
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China; Department of Neonatology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang, China; Key Laboratory of Perinatal Medicine of Wenzhou, Wenzhou, Zhejiang, China.
| |
Collapse
|
8
|
Chen J, Zhou Q, Cao Y, Tang X, Zhang Y, Wang L, Li J, Liang B. Identification of the potential role of PANoptosis-related genes in burns via bioinformatic analyses and experimental validation. Burns 2025; 51:107477. [PMID: 40233527 DOI: 10.1016/j.burns.2025.107477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 03/13/2025] [Accepted: 03/28/2025] [Indexed: 04/17/2025]
Abstract
BACKGROUND The treatment of burns is highly challenging due to their complex pathophysiological mechanisms. PANoptosis, as an important form of cell death, is suggested to play a crucial role in the inflammatory response and tissue damage following burns. However, the role of PANoptosis-related biomarkers in the pathophysiological processes of burns remains unclear. In this study, we aim to identify PANoptosis-related signature genes and validate them as biomarkers in burns METHODS: Burn-related datasets were obtained from the Gene Expression Omnibus(GEO) database. GSE37069 was used for bioinformatic analysis and machine learning, while GSE19743 was used specifically for external validation. A set of PANoptosis-associated genes was obtained from the GeneCards database. Three machine learning models (LASSO, RF, and SVM-RFE) and WGCNA were utilized to screen for signature genes. The diagnostic efficacy of the identified genes was assessed through receiver operating characteristic (ROC) curves. Gene Set Enrichment Analysis (GSEA) was performed to identify pathways associated with the signature genes, while single-sample gene set enrichment analysis (ssGSEA) was employed to investigate the immune landscape. Finally, Western blotting and RT-qPCR were employed to validate the signature genes. RESULTS BCL-2, CCAR1, CERK, TRIAP1, S100A8, and SNHG1 were identified as signature genes. The biological processes involving these genes mainly include endocytosis, apoptosis, and ECM receptor interaction. Immune infiltration analysis revealed that neutrophils, eosinophils, M0 macrophages, and monocytes are significantly elevated in burn samples. Additionally, these signature genes showed significant correlations with multiple immune cell types. Finally, Western blotting and RT-qPCR analysis revealed that the expression levels of BCL2, CCAR1, CERK, and TRIAP1 were significantly down-regulated in the burn groups compared to the normal groups, with the exception of S100A8. CONCLUSION Our study has identified BCL-2, CCAR1, CERK, and TRIAP1 as reliable potential biomarkers for burn injuries. These genes play crucial roles in immune response, wound healing, and anti-apoptotic mechanisms, which are key pathological processes involved in the progression of burn injuries. Specifically, BCL-2, CCAR1, CERK, and TRIAP1 have been shown to significantly impact the regulation of inflammation, the efficiency of wound repair, and the prevention of cell apoptosis during burn injury.
Collapse
Affiliation(s)
- Jiacong Chen
- Department of Anesthesiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou 510220, China
| | - Qin Zhou
- Department of Anesthesiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China
| | - Yang Cao
- Department of Anesthesiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou 510220, China
| | - Xuexian Tang
- Department of Anesthesiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou 510220, China
| | - Yan Zhang
- Department of Anesthesiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou 510220, China
| | - Lin Wang
- Department of Anesthesiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou 510220, China
| | - Junxi Li
- Department of Anesthesiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou 510220, China
| | - Bing Liang
- Department of Anesthesiology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou 510220, China.
| |
Collapse
|
9
|
Pu Y, Zhou Y, Guo T, Chai X, Yang G. PANoptosis-related gene biomarkers in aortic dissection. Arch Biochem Biophys 2025; 768:110385. [PMID: 40086567 DOI: 10.1016/j.abb.2025.110385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/17/2025] [Accepted: 03/11/2025] [Indexed: 03/16/2025]
Abstract
INTRODUCTION Programmed cell death of vascular smooth muscle cells (VSMCs) is critical in the pathogenesis of aortic dissection (AD), yet the role of PANoptosis-comprising pyroptosis, apoptosis, and necroptosis-remains unclear. METHODS We utilized the GSE213740 single-cell sequencing dataset to assess PANoptosis levels in VSMCs. Datasets GSE153434 and GSE147026 were employed to identify differentially expressed genes (DEGs) and perform weighted gene co-expression network analysis. PANoptosis gene sets were sourced from the GSEA website, with GSE52093 serving as the validation cohort. Gene Ontology, Kyoto Encyclopedia of Genes and Genomes (KEGG), and Protein-Protein Interaction analyses were conducted, along with assessments of upstream regulators and immune cell infiltration. Validation was performed on aortic tissues from AD patients and mouse models. RESULTS The single-cell dataset revealed an increased PANoptosis score in VSMCs in AD. Nineteen PANoptosis-related DEGs (PANDEGs) were identified, contributing to VSMC differentiation, DNA damage response, and apoptosis. KEGG analysis highlighted the P53 and TGF-β pathways, with PANDEGs positively correlating with immune cell infiltration. Key PANDEGs GADD45B, CDKN1A, and SOD2 were validated, showing co-expression with α-SMA. CONCLUSION The increased PANoptosis score in VSMCs suggests that GADD45B, CDKN1A, and SOD2 play crucial roles in AD.
Collapse
Affiliation(s)
- Yuting Pu
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China; Emergency Medicine and Difficult Disease Institute, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Yang Zhou
- Department of Intensive Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Tuo Guo
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China; Emergency Medicine and Difficult Disease Institute, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Xiangping Chai
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China; Emergency Medicine and Difficult Disease Institute, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
| | - Guifang Yang
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China; Emergency Medicine and Difficult Disease Institute, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
| |
Collapse
|
10
|
Zhao Y, Liang L, Jeon JE, Keshavjee S, Liu M. Ischemia/Reperfusion Upregulates Genes Related to PANoptosis in Human Lung Transplants. Transplantation 2025; 109:1004-1015. [PMID: 39528335 DOI: 10.1097/tp.0000000000005268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
BACKGROUND Activation of multiple programmed cell death (PCD) pathways has been reported in cellular and animal studies of ischemia/reperfusion injury in lung transplantation. However, the status of these pathways in human lung transplants remains unknown. This study investigates the involvement of PCD pathways and their relationship with inflammation and signaling pathways in human lung transplants. METHODS Transcriptomic analysis was conducted on 54 paired human lung tissue samples at the end of cold preservation time and 2 h after reperfusion, collected between 2008 and 2011. Gene Set Enrichment Analysis (GSEA) and single-sample GSEA were used to examine the activation of genes in 6 PCD pathways. The relationships between PCD pathways and inflammation, as well as signaling pathways, were assessed via single-gene GSEA. RESULTS GSEA results indicated that apoptosis and necroptosis were significantly upregulated after reperfusion in human lung transplants, whereas the gene sets related to pyroptosis, ferroptosis, autophagy, and cuproptosis were not significantly upregulated. Notably, single-sample GSEA demonstrated an intricate interplay among pyroptosis, apoptosis, and necroptosis, collectively referred to as PANoptosis, which is further supported by enrichment of genes related to PANoptosome, inflammatory response, and nuclear factor-κB and interferon signaling pathways, via single-gene GSEA assays. CONCLUSIONS This study demonstrated the genes of PANoptosis are upregulated in human lung grafts during reperfusion. The discovery of PANoptosis as an underlying mechanism of cell death in human lung grafts implies that effective therapeutics to prevent or reduce PANoptosis may alleviate ischemia/reperfusion injury and improve clinical lung transplant outcomes.
Collapse
Affiliation(s)
- Yajin Zhao
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Lubiao Liang
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Department of Thoracic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Jamie E Jeon
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Shaf Keshavjee
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Mingyao Liu
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
11
|
Bai Q, Wang C, Ding N, Wang Z, Liu R, Li L, Piao H, Song Y, Yan G. Eupalinolide B targets DEK and PANoptosis through E3 ubiquitin ligases RNF149 and RNF170 to negatively regulate asthma. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156657. [PMID: 40120540 DOI: 10.1016/j.phymed.2025.156657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/27/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
PURPOSE We investigated the mechanism by which eupalinolide B (EB) regulates DEK protein ubiquitination and degradation, and its impact on DEK-mediated receptor-interacting protein kinase 1 (RIPK)-PANoptosis pathway in allergic asthma. STUDY DESIGN AND METHODS In vitro studies were conducted on human bronchial epithelial cells (BEAS-2B) treated with EB and human-recombinant DEK. Mass spectrometry analysis, RNA sequencing, molecular docking, and functional assays were used to assess the interactions and effects of EB, DEK, and ring finger protein 149 and 170 (RNF149 and RNF170). In vivo experiments involved a house dust mite-induced asthma model in mice and evaluation of airway inflammation, DEK expression, and PANoptosis markers. RESULTS In vitro, EB could bind to DEK. RNF149 and RNF170 were identified as regulatory factors of DEK, polyubiquitinating the K349 site in the DEK coding DNA sequence region 270-350 through K48 linkages and leading to its degradation. RNA sequencing showed that DEK overexpression upregulated the expression of genes such as RIPK1, FADD, and Caspase 8. Treatment with DEK siRNA or EB reduced the activation of the RIPK1-PANoptosis pathway in BEAS-2B-DEK cells. In vivo, EB significantly reduced the levels of DEK in house dust mite-induced mice and alleviated pulmonary inflammatory cell infiltration, goblet cell hyperplasia, collagen fiber deposition, and eosinophil proportion in BALF. Knocking out the DEK gene reduced RIPK1-induced PANoptosis, and inhibited airway inflammation and cell apoptosis. CONCLUSION EB promotes the degradation of DEK by RNF149 and RNF170, inhibits the RIPK1-PANoptosis pathway, and may effectively suppress asthma. EB may become a potential drug for treating airway inflammation in asthma.
Collapse
Affiliation(s)
- Qiaoyun Bai
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji 133002, PR China; Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji 133002, PR China
| | - Chongyang Wang
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji 133002, PR China; Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji 133002, PR China
| | - Ningpo Ding
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji 133002, PR China; Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji 133002, PR China
| | - Zhiguang Wang
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji 133002, PR China; Department of Respiratory Medicine, Affiliated Hospital of Yanbian University, Yanji 133000, PR China
| | - Ruobai Liu
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji 133002, PR China; Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji 133002, PR China
| | - Liangchang Li
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji 133002, PR China; Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji 133002, PR China
| | - Hongmei Piao
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji 133002, PR China; Department of Respiratory Medicine, Affiliated Hospital of Yanbian University, Yanji 133000, PR China
| | - Yilan Song
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji 133002, PR China; Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji 133002, PR China; Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji 133002, PR China.
| | - Guanghai Yan
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji 133002, PR China; Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji 133002, PR China; Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji 133002, PR China.
| |
Collapse
|
12
|
He L, Zheng S, Zhan F, Lin N. The role of necroptosis in pathological pregnancies: Mechanisms and therapeutic opportunities. J Reprod Immunol 2025; 169:104460. [PMID: 40023097 DOI: 10.1016/j.jri.2025.104460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 02/02/2025] [Accepted: 02/19/2025] [Indexed: 03/04/2025]
Abstract
Necroptosis, a distinctive form of programmed cell death differs mechanistically from apoptosis pyroptosis, and autophagy, is characterized by the activation of receptor-interacting protein kinases (RIPK1/RIPK3) and their downstream effector, mixed lineage kinase domain-like protein (MLKL). This programmed cell death pathway serves as a crucial mediator of inflammatory responses and has been implicated in the pathogenesis of diverse pathological conditions. Recent evidence has implicated dysregulated necroptosis in the pathogenesis of severe pregnancy complications, including preeclampsia (PE), fetal growth restriction (FGR), recurrent spontaneous abortion (RSA), and gestational diabetes mellitus (GDM). In these disorders, necroptosis promotes placental dysfunction through multiple interconnected mechanisms: amplification of pro-inflammatory cytokine cascades, aberrant immune activation, disruption of plasma membrane integrity, and subsequent tissue injury.These pregnancy-related pathologies consistently demonstrate elevated necroptotic signatures, correlating with adverse maternal-fetal outcomes. This comprehensive review synthesizes current understanding of the molecular mechanisms underlying necroptosis, with particular emphasis on its pivotal role in the etiopathogenesis of pregnancy-related disorders. Furthermore, we critically evaluate the therapeutic potential of targeting the necroptotic signaling axis, providing novel perspectives for developing targeted interventions to improve clinical outcomes in complicated pregnancies.
Collapse
Affiliation(s)
- Lidan He
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350004, China; College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350122, China.
| | - Shan Zheng
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian 350004, China; College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350122, China
| | - Feng Zhan
- College of Engineering, Fujian Jiangxia University, Fuzhou 350108, China; School of Electronic Information Engineering, Taiyuan University of Science and Technology, Taiyuan, Shanxi 030024, China
| | - Na Lin
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350122, China; Medical Genetic Diagnosis and Therapy Center, Fujian Maternity and Child Health Hospital, Fuzhou 350122, China.
| |
Collapse
|
13
|
She H, Zheng J, Zhao G, Du Y, Tan L, Chen ZS, Wu Y, Li Y, Liu Y, Sun Y, Hu Y, Zuo D, Mao Q, Liu L, Li T. Arginase 1 drives mitochondrial cristae remodeling and PANoptosis in ischemia/hypoxia-induced vascular dysfunction. Signal Transduct Target Ther 2025; 10:167. [PMID: 40425583 PMCID: PMC12117058 DOI: 10.1038/s41392-025-02255-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 04/18/2025] [Accepted: 04/27/2025] [Indexed: 05/29/2025] Open
Abstract
Ischemic/hypoxic injury significantly damages vascular function, detrimentally impacting patient outcomes. Changes in mitochondrial structure and function are closely associated with ischemia/hypoxia-induced vascular dysfunction. The mechanism of this process remains elusive. Using rat models of ischemia and hypoxic vascular smooth muscle cells (VSMCs), we combined transmission electron microscopy, super-resolution microscopy, and metabolic analysis to analyze the structure and function change of mitochondrial cristae. Multi-omics approaches revealed arginase 1 (Arg1) upregulation in ischemic VSMCs, confirmed by in vivo and in vitro knockout models showing Arg1's protective effects on mitochondrial cristae, mitochondrial and vascular function, and limited the release of mtDNA. Mechanistically, Arg1 interacting with Mic10 led to mitochondrial cristae remodeling, together with hypoxia-induced VDAC1 lactylation resulting in the opening of MPTP and release of mtDNA of VSMCs. The released mtDNA led to PANoptosis of VSMCs via activation of the cGAS-STING pathway. ChIP-qPCR results demonstrated that lactate-mediated Arg1 up-regulation was due to H3K18la upregulation. VSMCs targeted nano-material PLGA-PEI-siRNA@PM-α-SMA (NP-siArg1) significantly improved vascular dysfunction. This study uncovers a new mechanism of vascular dysfunction following ischemic/hypoxic injury: a damaging positive feedback loop mediated by lactate-regulated Arg1 expression between the nucleus and mitochondria, leading to mitochondria cristae disorder and mtDNA release, culminating in VSMCs PANoptosis. Targeting VSMCs Arg1 inhibition offers a potential therapeutic strategy to alleviate ischemia/hypoxia-induced vascular impairments.
Collapse
Affiliation(s)
- Han She
- Department of Anesthesiology, Daping Hospital, Army Medical University, Chongqing, 400042, China
- Shock and Transfusion Department, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Jie Zheng
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Guozhi Zhao
- Department of Urology Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yunxia Du
- Department of Anesthesiology, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Lei Tan
- Department of Anesthesiology, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA
| | - Yinyu Wu
- Department of Anesthesiology, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Yong Li
- Department of Anesthesiology, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Yiyan Liu
- Shock and Transfusion Department, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Yue Sun
- Department of Anesthesiology, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Yi Hu
- Department of Anesthesiology, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Deyu Zuo
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Chongqing University of Chinese Medicine, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400021, China.
- Department of Research and Development, Chongqing Precision Medical Industry Technology Research Institute, Chongqing, 400000, China.
| | - Qingxiang Mao
- Department of Anesthesiology, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| | - Liangming Liu
- Shock and Transfusion Department, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| | - Tao Li
- Shock and Transfusion Department, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| |
Collapse
|
14
|
Qi L, Lan B, Zhao Z, Ma Y, Song J, Jia Q, Zhao P, Du X. Research advances of PANoptosis in gastrointestinal tumors. Int Immunopharmacol 2025; 159:114931. [PMID: 40414073 DOI: 10.1016/j.intimp.2025.114931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 05/08/2025] [Accepted: 05/19/2025] [Indexed: 05/27/2025]
Abstract
Gastric and colorectal cancers are acknowledged as the predominant types of gastrointestinal malignancies, significantly impacting the global cancer burden. Despite advancements in basic and clinical research on gastrointestinal cancer, the pathophysiological mechanisms and developmental processes underlying these diseases remain incompletely understood. The dysregulation of programmed cell death (PCD) has been identified as a crucial factor in the progression and metastasis of malignant tumors. The effective induction of cancer cell death continues to present a major challenge in contemporary cancer research. PANoptosis, a distinctive form of PCD integrating apoptosis, pyroptosis, and necroptosis, was introduced in 2019. Upon detecting relevant stimuli, PANoptosis sensors recruit key molecules from the three death modalities through domain-specific interactions to form a PANoptosome, which executes cell death. Recent discoveries suggest that PANoptosis plays a pivotal role in the development, progression, and drug resistance of gastrointestinal cancer. Enhancing PANoptosis will provide superior control over gastrointestinal tumors through multi-pathway crosstalk and inflammatory microenvironment modulation. This review aims to serve as a comprehensive resource for researchers by exploring the molecular foundation of PANoptosis, emphasizing its importance in gastrointestinal tumor development, and addressing current challenges as well as potential future research directions in this field.
Collapse
Affiliation(s)
- Lin Qi
- Department of General Surgery, First Medical Center of the Chinese PLA General Hospital, Beijing 100853, China; School of Medicine, Nankai University, Tianjin 300071, China
| | - Bin Lan
- Department of General Surgery, First Medical Center of the Chinese PLA General Hospital, Beijing 100853, China; Medical School of Chinese PLA, Beijing 100853, China
| | - Zhenting Zhao
- College of Life Science, Xinyang Normal University, Xinyang 464000, China
| | - Yizhao Ma
- Department of General Surgery, First Medical Center of the Chinese PLA General Hospital, Beijing 100853, China; School of Medicine, Nankai University, Tianjin 300071, China
| | - Jiachun Song
- Department of General Surgery, First Medical Center of the Chinese PLA General Hospital, Beijing 100853, China
| | - Qingzhe Jia
- Department of General Surgery, First Medical Center of the Chinese PLA General Hospital, Beijing 100853, China
| | - Pengyue Zhao
- Department of General Surgery, First Medical Center of the Chinese PLA General Hospital, Beijing 100853, China.
| | - Xiaohui Du
- Department of General Surgery, First Medical Center of the Chinese PLA General Hospital, Beijing 100853, China.
| |
Collapse
|
15
|
Guan J, Shi X, Ma J, Yin Y, Song G, Li Y, Chen X, Yan Y, Wang D, Liu S, Liu G, Zheng M, Ma F. Circular RNA-OGDH Promotes PANoptosis in Diabetic Cardiomyopathy: A Novel Mechanistic Insight. J Biol Chem 2025:110280. [PMID: 40412523 DOI: 10.1016/j.jbc.2025.110280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Revised: 04/29/2025] [Accepted: 05/07/2025] [Indexed: 05/27/2025] Open
Abstract
Diabetic cardiomyopathy (DCM) is a myocardial structural and functional abnormality directly caused by diabetes and is a principal factor in the development of cardiovascular complications in patients with diabetes. The study aims to investigate the role of circOGDH in the development of DCM and elucidate its precise underlying mechanisms. We established two well-characterised diabetic mouse models, C57BL/6J and db/db, and assessed cardiac function by serum lactate dehydrogenase activity assay and echocardiography, as well as quantitative histological analyses of the extent of myocardial fibrosis in combination with HE staining and Masson trichrome staining. The results demonstrated that there was a significant upregulation of circOGDH expression levels in myocardial tissues of mice in a diabetic state, accompanied by increased expression of key effector proteins of PANoptosis. It is noteworthy that the knockdown of circOGDH led to a substantial enhancement in cardiac function indices, a reduction in the area of myocardial fibrosis, and the effective inhibition of the PANoptosis process in myocardial tissues. In the H9c2 cells model, silencing of circOGDH also exhibited significant protective effects, including increased cell survival, reduced levels of oxidative stress, decreased apoptosis, and suppressed expression of PANoptosis-related proteins. Subsequent employing RNA pull-down, RNA immunoprecipitation and co-immunoprecipitation experimental methods have elucidated, for the first time, the molecular mechanism by which circOGDH specifically targets and regulates RIPK3 through the HMGB1 signalling pathway. The present study definitively demonstrated that up-regulation of circOGDH expression in a diabetic state could exacerbate pathological damage in diabetic cardiomyopathy by activating the HMGB1/RIPK3 signalling pathway.
Collapse
Affiliation(s)
- Jingyue Guan
- Department of Cardiology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, Hebei, 050031, China
| | - Xiaocui Shi
- Department of Cardiology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, Hebei, 050031, China
| | - Jianwei Ma
- Gastrointestinal Disease Diagnosis and Treatment Center, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, Hebei, 050031, China
| | - Yajuan Yin
- Department of Cardiology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, Hebei, 050031, China
| | - Guoyuan Song
- Department of Cardiology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, Hebei, 050031, China
| | - Yichen Li
- Department of Cardiology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, Hebei, 050031, China
| | - Xinyue Chen
- Department of Cardiology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, Hebei, 050031, China
| | - Yan Yan
- Department of Cardiology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, Hebei, 050031, China
| | - Dongxia Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Hebei Medical University, Shijiazhuang, Hebei, 050017, China
| | - Shangyu Liu
- Department of Cardiology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, Hebei, 050031, China
| | - Gang Liu
- Department of Cardiology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, Hebei, 050031, China; Hebei Key Laboratory of Cardiac Injury Repair Mechanism Study, Shijiazhuang, Hebei, 050031, China
| | - Mingqi Zheng
- Department of Cardiology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, Hebei, 050031, China; Hebei Key Laboratory of Heart and Metabolism, Shijiazhuang, Hebei, 050031, China.
| | - Fangfang Ma
- Department of Cardiology, The First Hospital of Hebei Medical University, 89 Donggang Road, Shijiazhuang, Hebei, 050031, China.
| |
Collapse
|
16
|
Ma C, Zhou X, Pan S, Liu L. AIM2 mediated neuron PANoptosis plays an important role in diabetes cognitive dysfunction. Behav Brain Res 2025; 491:115651. [PMID: 40404017 DOI: 10.1016/j.bbr.2025.115651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 04/19/2025] [Accepted: 05/20/2025] [Indexed: 05/24/2025]
Abstract
The increasing global aging population has led to a rise in diabetic cognitive dysfunction (DCD), a common complication of diabetes that significantly impacts the health of elderly individuals. Neuronal death is a key factor in cognitive impairment, with studies showing interactions between cellular pyroptosis, apoptosis, and necroptosis in the development of neurodegenerative disorders. This has led to the concept of PANoptosis, where these pathways work together to cause cell death. High glucose levels can induce neuronal damage and cognitive dysfunction in rats, leading to various forms of programmed cell death. It is hypothesized that high glucose can trigger neuronal PANoptosis, resulting in cognitive dysfunction. AIM2, an upstream regulator of PANoptosis, is closely associated with the pathogenesis of DCD. In DCD, dysregulated glucose metabolism induces the release of mitochondrial DNA (mtDNA), which acts as a ligand to activate the cell membrane-bound DNA sensor AIM2. Upon activation, AIM2 oligomerizes and recruits a caspase recruit domain (ASC), forming a complex that activates caspase-1. Caspase-1 activation subsequently triggers the production of pro-inflammatory cytokines, induces pyroptosis, and mediates apoptosis, necroptosis, and PANoptosis in neurons through signaling crosstalk. Understanding the pathophysiological mechanism of AIM2-mediated neuronal PANoptosis in DCD development can aid in early diagnosis and identify new therapeutic targets.
Collapse
Affiliation(s)
- Chengning Ma
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Hunan 4120208, China
| | - Xiang Zhou
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Hunan 4120208, China
| | - Siyang Pan
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Hunan 4120208, China
| | - Lumei Liu
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Hunan 4120208, China.
| |
Collapse
|
17
|
Wang H, Feng X, He H, Li L, Wen Y, Liu X, He B, Hua S, Sun S. Crosstalk between autophagy and other forms of programmed cell death. Eur J Pharmacol 2025; 995:177414. [PMID: 39986593 DOI: 10.1016/j.ejphar.2025.177414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/13/2025] [Accepted: 02/19/2025] [Indexed: 02/24/2025]
Abstract
Cell death occurs continuously throughout individual development. By removing damaged or senescent cells, cell death not only facilitates morphogenesis during the developmental process, but also contributes to maintaining homeostasis after birth. In addition, cell death reduces the spread of pathogens by eliminating infected cells. Cell death is categorized into two main forms: necrosis and programmed cell death. Programmed cell death encompasses several types, including autophagy, pyroptosis, apoptosis, necroptosis, ferroptosis, and PANoptosis. Autophagy, a mechanism of cell death that maintains cellular equilibrium via the breakdown and reutilization of proteins and organelles, is implicated in regulating almost all forms of cell death in pathological contexts. Notably, necroptosis, ferroptosis, and PANoptosis are directly classified as autophagy-mediated cell death. Therefore, regulating autophagy presents a therapeutic approach for treating diseases such as inflammation and tumors that arise from abnormalities in other forms of programmed cell death. This review focuses on the crosstalk between autophagy and other programmed cell death modalities, providing new perspectives for clinical interventions in inflammatory and neoplastic diseases.
Collapse
Affiliation(s)
- Huaiyuan Wang
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China; Clinical Medicine, class 3, 2022 Grade, Kunming Medical University, Kunming, China
| | - Xiran Feng
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China; Clinical Medicine, Kunming Medical University-Shanghai Jiaotong University Joint Program, 2022 Grade, Kunming Medical University, Kunming, China
| | - Huilin He
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Lingyu Li
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Yiqiong Wen
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Xiaofei Liu
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Bifeng He
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Shu Hua
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China
| | - Shibo Sun
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital, Kunming Medical University, Kunming, China.
| |
Collapse
|
18
|
Wu Q, Qi S, Kang Z, Bai X, Li Z, Cheng J, Dong X. PANoptosis in Sepsis: A Central Role and Emerging Therapeutic Target. J Inflamm Res 2025; 18:6245-6261. [PMID: 40386177 PMCID: PMC12085136 DOI: 10.2147/jir.s513367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 05/04/2025] [Indexed: 05/20/2025] Open
Abstract
The pathogenesis of sepsis is intricately linked to regulated cell death. As a novel form of regulated cell death, PANoptosis plays a critical role in driving the inflammatory response, impairing immune cell function, and contributing to multi-organ dysfunction in sepsis. This review explores the molecular mechanisms underlying PANoptosis and its involvement in sepsis. By activating multiple pathways, PANoptosis promotes the release of inflammatory cytokines, triggering a cytokine storm that disrupts immune cell homeostasis and exacerbates organ damage. Emerging therapeutic strategies targeting PANoptosis, including chemotherapeutic agents and herbal remedies, are showing potential for clinical application. The concept of targeting PANoptosis offers a promising avenue for developing innovative treatments for sepsis.
Collapse
Affiliation(s)
- Qiqi Wu
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Siyuan Qi
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Zhaofeng Kang
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Xiangjun Bai
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Zhanfei Li
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Jing Cheng
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Xijie Dong
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| |
Collapse
|
19
|
Chen Y, You Y, Xie Y, Li X, Zhu Z, Li W, Du X, Yan Z. ZBP1 synchronized with periodontopathogenesis as the essential pattern recognition receptor. Microb Pathog 2025; 205:107678. [PMID: 40349992 DOI: 10.1016/j.micpath.2025.107678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 05/03/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025]
Abstract
BACKGROUND Periodontitis is a chronic inflammatory disease impacting quality of life. Understanding its pathogenesis is key to developing effective treatments. This study aimed to identify key pattern recognition receptors (PRRs) involved in periodontitis and elucidate their roles in disease progression. METHODS Periodontal tissues from healthy individuals and those with periodontitis were analyzed using RNA-sequencing, quantitative real-time PCR(qRT-PCR), and immunohistochemical analysis. Paired tissues collected before and after non-surgical treatment were analyzed via 4D-microDIA proteomics and Western blot. RESULTS RNA-sequencing showed significantly higher expression of Z-DNA binding protein 1(ZBP1) and absent in melanoma 2(AIM2) in periodontitis tissues compared to healthy controls, confirmed by qRT-PCR. Post-treatment proteomics indicated significant downregulation of ZBP1, with a non-significant trend for AIM2. Immunohistochemical staining localized ZBP1 to the middle and superficial layers of the gingival epithelium and around deep pockets in periodontitis, while AIM2 was detected in the junctional epithelium and extended throughout the pocket epithelium in periodontitis. CONCLUSIONS ZBP1 is highlighted as a key PRR in periodontitis, with significant regulatory potential. AIM2 may play a secondary role. Their distinct spatial distributions suggest involvement in specific microenvironments within periodontal tissues, mediating responses to microbial and inflammatory challenges. ZBP1 may be a critical receptor initiating periodontitis.
Collapse
Affiliation(s)
- Yu Chen
- Department of Dentistry, People's Hospital of Longhua, Shenzhen, 518109, China; Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China.
| | - Yuehua You
- Department of Dentistry, People's Hospital of Longhua, Shenzhen, 518109, China.
| | - Yi Xie
- Department of Pathology, People's Hospital of Longhua, Shenzhen, 518109, China.
| | - Xiaoyu Li
- Department of Dentistry, People's Hospital of Longhua, Shenzhen, 518109, China.
| | - Zhigao Zhu
- Department of Dentistry, People's Hospital of Longhua, Shenzhen, 518109, China.
| | - Wenlong Li
- Department of Dentistry, People's Hospital of Longhua, Shenzhen, 518109, China.
| | - Xinya Du
- Department of Dentistry, People's Hospital of Longhua, Shenzhen, 518109, China.
| | - Zhengbin Yan
- Department of Dentistry, People's Hospital of Longhua, Shenzhen, 518109, China.
| |
Collapse
|
20
|
Huang Y, Qian J, Luan Z, Han J, Tang L. Comprehensive Analysis Reveals the Molecular Features and Immune Infiltration of PANoptosis-Related Genes in Metabolic Dysfunction-Associated Steatotic Liver Disease. BIOLOGY 2025; 14:518. [PMID: 40427707 PMCID: PMC12108815 DOI: 10.3390/biology14050518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 05/04/2025] [Accepted: 05/06/2025] [Indexed: 05/29/2025]
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD), a chronic inflammatory disorder characterized by alcohol-independent hepatic lipid accumulation, remains poorly understood in terms of PANoptosis involvement. METHODS We integrated high-throughput sequencing data with bioinformatics to profile differentially expressed genes (DEGs) and immune infiltration patterns in MASLD, identifying PANoptosis-associated DEGs (PANoDEGs). Machine learning algorithms prioritized key PANoDEGs, while ROC curves assessed their diagnostic efficacy. Cellular, animal, and clinical validations confirmed target expression. RESULTS Three PANoDEGs (SNHG16, Caspase-6, and Dynamin-1-like protein) exhibited strong MASLD associations and diagnostic significance. Immune profiling revealed elevated M1 macrophages, naïve B cells, and activated natural killer cells in MASLD tissues versus controls. Further experiments verified the expression of the key PANoDEGs. CONCLUSIONS This study provides new insights for further studies on the pathogenesis and treatment strategies of PANoptosis in MASLD.
Collapse
Affiliation(s)
- Yan Huang
- Medical College, Yangzhou University, Yangzhou 225000, China
| | - Jingyu Qian
- Taizhou School of Clinical Medicine, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou 225300, China
| | - Zhengyun Luan
- Taizhou School of Clinical Medicine, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou 225300, China
| | - Junling Han
- Taizhou School of Clinical Medicine, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou 225300, China
| | - Limin Tang
- Taizhou School of Clinical Medicine, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou 225300, China
| |
Collapse
|
21
|
Xuan X, Fan J, Zhang J, Ren M, Feng L. Immune in myocardial ischemia/reperfusion injury: potential mechanisms and therapeutic strategies. Front Immunol 2025; 16:1558484. [PMID: 40406107 PMCID: PMC12094985 DOI: 10.3389/fimmu.2025.1558484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 04/14/2025] [Indexed: 05/26/2025] Open
Abstract
Myocardial infarction (MI), which is characterized by high morbidity and mortality, is a serious threat to human life and health, and timely reperfusion therapy to save ischemic myocardium is currently the most effective intervention. Although reperfusion therapy effectively restores coronary blood flow and maximally limits the infarct size, it triggers additional cell death and tissue damage, which is known as myocardial ischemia/reperfusion injury (MIRI). Multiple immune cells are present in the reperfusion area, executing specific functions and engaging in crosstalk during diverse stages, constituting a complex immune microenvironment involved in tissue repair and regeneration after MIRI. Immunotherapy brings new hope for treating ischemic heart disease by modulating the immune microenvironment. In this paper, we explore the regulatory roles of various immune cells during MIRI and the close relationship between different cell deaths and the immune microenvironment. In addition, we present the current status of research on targeting the immune system to intervene in MIRI, with the expectation of providing a basis for achieving clinical translation.
Collapse
Affiliation(s)
- Xiaoyu Xuan
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jilin Fan
- Department of Rehabilitation, The Affiliated Hospital of Binzhou Medical University, Binzhou, Shandong, China
| | - Jingyi Zhang
- Department of Traditional Chinese Medicine, The Second Affiliated Hospital of Shandong First Medical University, Shandong, Taian, China
| | - Ming Ren
- Baokang Hospital Affiliated to Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Limin Feng
- The Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Binhai New Area Traditional Chinese Medicine Hospital, Tianjin, China
| |
Collapse
|
22
|
Guo J, Meng S, Zhang J, Wang N, Guo F. Zn 2+ regulates mitochondrial DNA efflux to inhibit AIM2-mediated ZBP1-PANoptosome pathway and alleviate septic myocardial injury. Chem Biol Interact 2025; 417:111525. [PMID: 40348119 DOI: 10.1016/j.cbi.2025.111525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 04/18/2025] [Accepted: 04/22/2025] [Indexed: 05/14/2025]
Abstract
This study was performed to investigate the mechanism by which zinc ion regulated mitochondrial DNA (mtDNA) efflux to inhibit the AIM2-mediated ZBP1-PANoptosome pathway and alleviate sepsis-induced myocardial injury. Here we discovered that zinc ions suppressed mitochondrial DNA release, thereby protecting the heart from LPS-induced damage in mice. In addition, LPS induced mPTP opening and mediated mtDNA efflux in cardiomyocytes, which drove AIM2 activation and ZBP1-PANoptosome multiprotein complex formation, leading to pan-apoptotic cardiomyocyte death. Zn2+ prevented mPTP opening to inhibit mtDNA efflux-driven AIM2 and ZBP1-PANoptosome multiprotein complex formation and alleviate PANoptosis. Knockdown of AIM2 alleviated LPS-induced PANoptosis in cardiomyocytes. LPS-induced PANoptosis in cardiomyocytes by regulating the ZBP1/RIPK3 pathway. However, activation of the ZBP1/RIPK3 pathway partially reversed the inhibitory effect of Zn2+ on PANoptosis in cardiomyocytes. Taken together, Zn2+ regulated mitochondrial DNA efflux to inhibit the AIM2-mediated ZBP1-PANoptosome pathway to alleviate septic myocardial injury.
Collapse
Affiliation(s)
- Jun Guo
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu Province, PR China; Department of Critical Care Medicine, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430014, Hubei Province, PR China
| | - Shanshan Meng
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu Province, PR China
| | - Jin Zhang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu Province, PR China
| | - Ni Wang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu Province, PR China
| | - Fengmei Guo
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu Province, PR China.
| |
Collapse
|
23
|
Nadendla EK, Tweedell RE, Kasof G, Kanneganti TD. Caspases: structural and molecular mechanisms and functions in cell death, innate immunity, and disease. Cell Discov 2025; 11:42. [PMID: 40325022 PMCID: PMC12052993 DOI: 10.1038/s41421-025-00791-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 03/05/2025] [Indexed: 05/07/2025] Open
Abstract
Caspases are critical regulators of cell death, development, innate immunity, host defense, and disease. Upon detection of pathogens, damage-associated molecular patterns, cytokines, or other homeostatic disruptions, innate immune sensors, such as NLRs, activate caspases to initiate distinct regulated cell death pathways, including non-lytic (apoptosis) and innate immune lytic (pyroptosis and PANoptosis) pathways. These cell death pathways are driven by specific caspases and distinguished by their unique molecular mechanisms, supramolecular complexes, and enzymatic properties. Traditionally, caspases are classified as either apoptotic (caspase-2, -3, -6, -7, -8, -9, and -10) or inflammatory (caspase-1, -4, -5, and -11). However, extensive data from the past decades have shown that apoptotic caspases can also drive lytic inflammatory cell death downstream of innate immune sensing and inflammatory responses, such as in the case of caspase-3, -6, -7, and -8. Therefore, more inclusive classification systems based on function, substrate specificity, or the presence of pro-domains have been proposed to better reflect the multifaceted roles of caspases. In this review, we categorize caspases into CARD-, DED-, and short/no pro-domain-containing groups and examine their critical functions in innate immunity and cell death, along with their structural and molecular mechanisms, including active site/exosite properties and substrates. Additionally, we highlight the emerging roles of caspases in cellular homeostasis and therapeutic targeting. Given the clinical relevance of caspases across multiple diseases, improved understanding of these proteins and their structure-function relationships is critical for developing effective treatment strategies.
Collapse
Affiliation(s)
- Eswar Kumar Nadendla
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Rebecca E Tweedell
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Gary Kasof
- Cell Signaling Technology, Danvers, MA, USA
| | | |
Collapse
|
24
|
Qu C, Yang H. Prognostic Significance and Immune Environment Analysis Using PANoptosis Molecular Clustering in Gastric Cancer. Med Sci Monit 2025; 31:e947710. [PMID: 40317125 PMCID: PMC12057512 DOI: 10.12659/msm.947710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 02/27/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND Stomach adenocarcinoma (STAD) is a common malignant tumor, known for its poor prognosis and challenges in early detection. PANoptosis, a recently discovered form of cell death, is characterized by the integrated activation of pyroptosis, apoptosis, and/or necroptosis pathways. The connection between PANoptosis and the initiation, progression, and prognosis of gastric cancer remains inadequately investigated. MATERIAL AND METHODS Previous research has identified 19 PANoptosis-related genes (PRGs). Using these genes, we performed an in-depth analysis of gastric cancer to identify differentially expressed genes related to prognosis (PRDEGs). These differentially expressed genes were subsequently identified. We analyzed the risk scores, prognoses, and immune landscapes of the patients. Confirmed PRGs and gene clusters have been linked to cancer initiation and progression, patient survival, and immunity. Risk scores were computed, and patients were categorized into 2 groups on the basis of prognostic characteristics linked to 8 specific genes. To increase the accuracy of predicting patient survival, we developed a nomogram that integrates the risk score with various clinical characteristics. RESULTS The analysis revealed that gastric cancer patients classified into high-risk subgroups experienced reduced survival times and a diminished response to immunotherapy. We also found that risk scores demonstrated correlations with immune cell infiltration, tumor microenvironment characteristics (TME), and cancer stem cell (CSC) levels. The differential expression of GPA33 and APOD between gastric tumor and normal tissues was validated by RT-qPCR and immunohistochemical data from the Human Protein Atlas (HPA). In conclusion, our research indicates that genes linked to PANoptosis may serve as key indicators for evaluating the prognosis and survival rates of patients with gastric cancer. CONCLUSIONS This research has the potential to improve the early detection of gastric cancer and contribute to the development of more effective therapeutic approaches.
Collapse
|
25
|
Huang C, Li J, Wu R, Li Y, Zhang C. Targeting pyroptosis for cancer immunotherapy: mechanistic insights and clinical perspectives. Mol Cancer 2025; 24:131. [PMID: 40319304 PMCID: PMC12049004 DOI: 10.1186/s12943-025-02344-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Accepted: 04/26/2025] [Indexed: 05/07/2025] Open
Abstract
Pyroptosis is a distinct form of programmed cell death characterized by the rupture of the cell membrane and robust inflammatory responses. Increasing evidence suggests that pyroptosis significantly affects the tumor microenvironment and antitumor immunity by releasing damage-associated molecular patterns (DAMPs) and pro-inflammatory mediators, thereby establishing it as a pivotal target in cancer immunotherapy. This review thoroughly explores the molecular mechanisms underlying pyroptosis, with a particular focus on inflammasome activation and the gasdermin family of proteins (GSDMs). It examines the role of pyroptotic cell death in reshaping the tumor immune microenvironment (TIME) involving both tumor and immune cells, and discusses recent advancements in targeting pyroptotic pathways through therapeutic strategies such as small molecule modulators, engineered nanocarriers, and combinatory treatments with immune checkpoint inhibitors. We also review recent advances and future directions in targeting pyroptosis to enhance tumor immunotherapy with immune checkpoint inhibitors, adoptive cell therapy, and tumor vaccines. This study suggested that targeting pyroptosis offers a promising avenue to amplify antitumor immune responses and surmount resistance to existing immunotherapies, potentially leading to more efficacious cancer treatments.
Collapse
Affiliation(s)
- Chen Huang
- Department of Biotherapy, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Jiayi Li
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Ruiyan Wu
- West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yangqian Li
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Chenliang Zhang
- Division of Abdominal Tumor Multimodality Treatment, Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
26
|
Peng X, Gao Y, Liu J, Shi X, Li W, Ma Y, Li X, Li H. Mitochondria-derived vesicles: A promising and potential target for tumour therapy. Clin Transl Med 2025; 15:e70320. [PMID: 40356246 PMCID: PMC12069804 DOI: 10.1002/ctm2.70320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 04/10/2025] [Accepted: 04/16/2025] [Indexed: 05/15/2025] Open
Abstract
Mitochondria-derived vesicles (MDVs) participate in early cellular defence mechanisms initiated in response to mitochondrial damage. They maintain mitochondrial quality control (MQC) by clearing damaged mitochondrial components, thereby ensuring the normal functioning of cellular processes. This process is crucial for cell survival and health, as mitochondria are the energy factories of cells, and their damage can cause cellular dysfunction and even death. Recent studies have shown that MDVs not only maintain mitochondrial health but also have a significant impact on tumour progression. MDVs selectively encapsulate and transport damaged mitochondrial proteins under oxidative stress and reduce the adverse effects of mitochondrial damage on cells, which may promote the survival and proliferation of tumour cells. Furthermore, it has been indicated that after cells experience mild stress, the number of MDVs significantly increases within 2-6 h, whereas mitophagy, a process of clearing damaged mitochondria, occurs 12-24 h later. This suggests that MDVs play a critical role in the early stress response of cells. Moreover, MDVs also have a significant role in intercellular communication, specifically in the tumour microenvironment. They can carry and transmit various bioactive molecules, such as proteins, nucleic acids, and lipids, which regulate tumour cell's growth, invasion, and metastasis. This intercellular communication may facilitate tumour spread and metastasis, making MDVs a potential therapeutic target. Advances in MDV research have identified novel biomarkers, clarified regulatory mechanisms, and provided evidence for clinical use. These breakthroughs pave the way for novel MDV-targeted therapies, offering improved treatment alternatives for cancer patients. Further research can identify MDVs' role in tumour development and elucidate future cancer treatment horizons.
Collapse
Affiliation(s)
- Xueqiang Peng
- Department of General SurgeryThe Fourth Affiliated HospitalChina Medical UniversityShenyangChina
- Group of Chronic Disease and Environmental GenomicsSchool of Public HealthChina Medical UniversityShenyangChina
- Shenyang Clinical Medical Research Center for DiagnosisTreatment and Health Management of Early Digestive CancerShenyangChina
| | - Yu Gao
- Department of General SurgeryThe Fourth Affiliated HospitalChina Medical UniversityShenyangChina
- Shenyang Clinical Medical Research Center for DiagnosisTreatment and Health Management of Early Digestive CancerShenyangChina
| | - Jiaxing Liu
- Department of General SurgeryThe Fourth Affiliated HospitalChina Medical UniversityShenyangChina
- Shenyang Clinical Medical Research Center for DiagnosisTreatment and Health Management of Early Digestive CancerShenyangChina
| | - Xinxin Shi
- Department of General SurgeryThe First Hospital of Anhui University of Science & TechnologyHuainanChina
| | - Wei Li
- Department of General SurgeryThe First Hospital of Anhui University of Science & TechnologyHuainanChina
| | - Yingbo Ma
- Depatment of Hepatobiliary SurgeryAir Force Medical CenterBeijingChina
| | - Xuexin Li
- Department of General SurgeryThe Fourth Affiliated HospitalChina Medical UniversityShenyangChina
- Shenyang Clinical Medical Research Center for DiagnosisTreatment and Health Management of Early Digestive CancerShenyangChina
- Division of Genome BiologyDepartment of Medical Biochemistry and BiophysicsKarolinska InstitutetStockholmSweden
| | - Hangyu Li
- Department of General SurgeryThe Fourth Affiliated HospitalChina Medical UniversityShenyangChina
- Shenyang Clinical Medical Research Center for DiagnosisTreatment and Health Management of Early Digestive CancerShenyangChina
- Department of General SurgeryThe First Affiliated Hospital of Jinzhou Medical UniversityJinzhouLiaoningChina
| |
Collapse
|
27
|
Zhong L, Guo X, Deng L, Wang X, He H, Wu N, Tang R, Chen L, Chen Y, Li P. Dopant-Regulated Piezocatalysts Evoke Sonopiezoelectric and Enzymatic PANoptosis for Synergistic Cancer Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2500406. [PMID: 40056039 PMCID: PMC12061309 DOI: 10.1002/advs.202500406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 02/17/2025] [Indexed: 05/10/2025]
Abstract
Piezocatalyst-enabled sonopiezoelectric therapy offers noninvasive treatment with high spatiotemporal selectivity, yet existing piezocatalysts are limited by suboptimal efficacy, cancer cell resistance to oxidative stress, and biosafety concerns. Here, hafnia (HfO2), one of the only few FDA-approved inorganic nanomaterials for clinical trials, is identified as a promising piezocatalyst with high translational potential for sonopiezoelectric and enzymatic PANoptosis-boosted nanocatalytic therapy. Specifically, engineered transition metal-substituted HfO2 nanocatalysts are synthesized to optimize piezoelectric and enzyme-mimicking activities. Among these, Mn-substituted HfO2 with a 20% Mn ratio (HMO) demonstrates superior performance in sono-triggered reactive oxygen species generation, attributed to its reduced bandgap and increased oxygen vacancies. HMO also exhibits multiple enzyme-mimicking activities, including peroxidase (POD), catalase (CAT), and glutathione peroxidase (GPx), amplifying oxidative stress through tumor-specific catalytic reactions. These dual catalytic effects enable the activation of cancer cell PANoptosis to elicit a robust antitumor immune response. Biological evaluations show significant tumor suppression and antitumor immune responses by HMO-mediated nanocatalytic therapy. Unlike utilizing the radiosensitization ability of HfO2 in the clinic, this work unveils the distinctive sonopiezoelectric effect and multienzymatic activities of HfO2-based nanocatalysts for biomedical applications, holding the potential to overcome the challenges of radiation damage associated with radiotherapy.
Collapse
Affiliation(s)
- Linhong Zhong
- Ultrasound Department of the Second Affiliated Hospital of Chongqing Medical UniversityChongqing Key Laboratory of Ultrasound Molecular ImagingChongqing400010P. R. China
| | - Xun Guo
- Ultrasound Department of the Second Affiliated Hospital of Chongqing Medical UniversityChongqing Key Laboratory of Ultrasound Molecular ImagingChongqing400010P. R. China
| | - Liming Deng
- The First Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Xiaoting Wang
- Ultrasound Department of the Second Affiliated Hospital of Chongqing Medical UniversityChongqing Key Laboratory of Ultrasound Molecular ImagingChongqing400010P. R. China
| | - Hongye He
- Ultrasound Department of the Second Affiliated Hospital of Chongqing Medical UniversityChongqing Key Laboratory of Ultrasound Molecular ImagingChongqing400010P. R. China
| | - Nianhong Wu
- Ultrasound Department of the Second Affiliated Hospital of Chongqing Medical UniversityChongqing Key Laboratory of Ultrasound Molecular ImagingChongqing400010P. R. China
| | - Rui Tang
- Ultrasound Department of the Second Affiliated Hospital of Chongqing Medical UniversityChongqing Key Laboratory of Ultrasound Molecular ImagingChongqing400010P. R. China
| | - Liang Chen
- Materdicine LabSchool of Life SciencesShanghai UniversityShanghai200444P. R. China
| | - Yu Chen
- Materdicine LabSchool of Life SciencesShanghai UniversityShanghai200444P. R. China
- Shanghai Institute of MaterdicineShanghai200051P. R. China
| | - Pan Li
- Ultrasound Department of the Second Affiliated Hospital of Chongqing Medical UniversityChongqing Key Laboratory of Ultrasound Molecular ImagingChongqing400010P. R. China
| |
Collapse
|
28
|
Gupta J, Mohammed MH, Alghazali T, Uthirapathy S, R R, Thakur V, Kaur M, Naidu KS, Kubaev A, Al-Mukhtar MM. Inflammasomes and autophagy in cancer: unlocking targeted therapies. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04184-x. [PMID: 40310530 DOI: 10.1007/s00210-025-04184-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Accepted: 04/13/2025] [Indexed: 05/02/2025]
Abstract
This study clarifies the interaction between autophagy and inflammasome within the cancer framework. The inflammasome generates pro-inflammatory cytokines to direct the immune response to pathogens and cellular stressors. Autophagy maintains cellular homeostasis and can either promote or inhibit cancer. These pathways interact to affect tumorigenesis, immune responses, and therapy. Autophagy controls inflammasome activity by affecting cancer pathogenesis and tumor microenvironment inflammation, highlighting novel cancer therapeutic approaches. Recent studies indicate that modulating autophagy and inflammasome pathways can boost anti-cancer immunity, reduce drug-resistance, and improve therapeutic efficacy. Recent studies indicate modulating inflammasome and autophagy pathways can augment anti-cancer immunity, mitigate therapy resistance, and improve treatment efficacy. Cancer research relies on understanding the inflammasome-autophagy relationship to develop targeted therapies that enhance anti-tumor efficacy and reduce inflammatory symptoms. Customized therapies may improve outcomes based on autophagy gene variations and inflammasome polymorphisms. This study investigates autophagy pathways and the inflammasome in tumor immunopathogenesis, cytokine function, and cancer therapeutic strategies, highlighting their significance in cancer biology and treatment.
Collapse
Affiliation(s)
- Jitendra Gupta
- Institute of Pharmaceutical Research, GLA University, Mathura, Pin Code 281406, U.P., India.
| | - Mohammed Hashim Mohammed
- Medical Laboratory Techniques Department, College of Health and Medical Technology, Al-Maarif University, Anbar, Iraq.
| | | | - Subasini Uthirapathy
- Pharmacy Department, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Roopashree R
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Vishal Thakur
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| | - Manpreet Kaur
- Department of Pharmacy, Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, 140307, Punjab, India
| | - K Satyam Naidu
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra, Pradesh- 531162, India
| | - Aziz Kubaev
- Department of Maxillofacial Surgery, Samarkand State Medical University, 18 Amir Temur Street, 140100, Samarkand, Uzbekistan
| | - Mahmoud Mussleh Al-Mukhtar
- Anesthesia Techniques Department, College of Health and Medical Techniques, Al-Mustaqbal University, 51001, Babylon, Iraq
| |
Collapse
|
29
|
Tang C. Immunologic cell deaths: involvement in the pathogenesis and intervention therapy of periodontitis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:4957-4968. [PMID: 39718617 DOI: 10.1007/s00210-024-03732-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 12/12/2024] [Indexed: 12/25/2024]
Abstract
Periodontitis is one of the most common diseases and primary causes of tooth loss. The main factor that causes periodontitis is an overactive host immunological response. An in-depth investigation into the molecular pathways that cause periodontitis can aid in creating novel therapeutic approaches for periodontitis and its related systemic disorders. Several immunologic cell death (ICD) pathways have been implicated in advancing periodontitis. Nevertheless, there is still a substantial lack of understanding surrounding the precise molecular mechanisms of ICD in periodontitis. Additionally, the beneficial feature of ICD in periodontitis, which involves its ability to eliminate pathogens, needs further confirmation. According to this, a comprehensive literature search utilizing the Web of Science™, PubMed®, and Scopus® databases was conducted. Only items published in the English language up until October 2024 were taken into account, and finally, 65 relevant papers were selected to be included in this review. In this article, we present a comprehensive analysis of the processes and outcomes of ICD activation in the progression of periodontitis. Lastly, the present difficulties linked to ICDs as a viable treatment option for periodontitis are emphasized.
Collapse
Affiliation(s)
- Chenyao Tang
- Department of Stomatology, Yongzhou Central Hospital, Yongzhou, Hunan Province, 425000, China.
| |
Collapse
|
30
|
Liu Z, Chen DH, Lin ZH, Wang ZY, Peng H, Liu RT, Hu ZC, He YH, Wei XJ, Zhang CQ, Feng Y, Tang Q, Zhu ZZ. In-situ Sprayed platelet-derived small extracellular vesicles for the skin flap survival by reducing PANoptosis. Biomaterials 2025; 316:123001. [PMID: 39671720 DOI: 10.1016/j.biomaterials.2024.123001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 11/02/2024] [Accepted: 12/04/2024] [Indexed: 12/15/2024]
Abstract
Necrosis at the distal end of random skin flaps remains a significant challenge, limiting the clinical application of these flaps in plastic and reconstructive surgery. Inhibiting ischemia/reperfusion (I/R) injury and promoting the formation of neovascular networks are critical preventive strategies. Platelet-derived small extracellular vesicles (PL-sEV) are nanocarriers of growth factors that provide an alternative to clinically used platelet-rich plasma and platelet lysates, offering higher growth factor concentrations and lower immunogenicity. In this study, PANoptosis, a distinct form of inflammatory cell death, was fully characterized in a random skin flap model. Subcutaneous injection of PL-sEV improved ischemic skin flap survival by enhancing blood perfusion and reducing PANoptosis levels. In vitro, PL-sEV inhibited oxygen-glucose deprivation/reoxygenation-induced dysfunction in human umbilical vein endothelial cells. Furthermore, PL-sEV was incorporated into a thermosensitive triblock hydrogel, creating a sprayable delivery system (PLEL@PL-sEV). Mechanistic analysis through RNA sequencing indicated that the protective effects of PL-sEV against PANoptosis likely resulted from its anti-inflammatory properties, particularly via suppression of the NF-κB signaling pathway. This novel hydrogel system demonstrated controlled release of PL-sEV and proved effective in improving skin flap transplantation outcomes.
Collapse
Affiliation(s)
- Zhe Liu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - De-Heng Chen
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Zi-Hao Lin
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Zi-Yi Wang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Hao Peng
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Ruo-Tao Liu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Zhi-Chao Hu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Yao-Hua He
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Xiao-Juan Wei
- Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China.
| | - Chang-Qing Zhang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China
| | - Yong Feng
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China.
| | - Qian Tang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China.
| | - Zhen-Zhong Zhu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 600 Yishan Road, Shanghai, 200233, China.
| |
Collapse
|
31
|
Luo Q, Shen L, Yang S, Zhang Y, Pan Y, Wu Z, Shu Q, Chen Q. Caspase-1-licensed pyroptosis drives dsRNA-mediated necroptosis and dampens host defense against bacterial pneumonia. PLoS Pathog 2025; 21:e1013167. [PMID: 40359428 PMCID: PMC12121916 DOI: 10.1371/journal.ppat.1013167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/29/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025] Open
Abstract
Bacterial lung infections cause severe host responses. Here, we showed that global deficiency of caspase-1 can protect against lethal pulmonary Escherichia coli infection by reducing the necroptosis of infiltrated neutrophils, which are key players in immune responses in the lung. Mechanistically, neutrophil necroptosis was not directly triggered in a cell-intrinsic manner by invading bacteria but was triggered by bacteria-stimulated pyroptotic epithelial cell supernatants in vitro. In validation experiments, chimeric mice with nonhematopoietic caspase-1 or GSDMD knockout were protected from lung E. coli infection and exhibited decreased neutrophil death. Nonhematopoietic pyroptosis facilitates the release of dsRNAs and contributes to neutrophil ZBP1-related necroptosis. Moreover, blocking dsRNA or depleting ZBP1 ameliorated the pathophysiological process of pulmonary E. coli infection. Overall, our results demonstrate a paradigm of communication between necroptosis and pyroptosis in different cell types in cooperation with microbes and hosts and suggest that therapeutic targeting of the pyroptosis or necroptosis pathway may prevent pulmonary bacterial infection.
Collapse
Affiliation(s)
- Qinyu Luo
- Department of Clinical Research Center, The Children’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lihua Shen
- Department of Clinical Research Center, The Children’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shiyue Yang
- Department of Anesthesiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yan Zhang
- Department of Clinical Research Center, The Children’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yihang Pan
- Department of Clinical Research Center, The Children’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zehua Wu
- Department of Clinical Research Center, The Children’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiang Shu
- Department of Clinical Research Center, The Children’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qixing Chen
- Department of Clinical Research Center, The Children’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
32
|
Kumar SP, Nadendla EK, Malireddi RKS, Haque SA, Mall R, Neuwald AF, Kanneganti TD. Evolutionary and Functional Analysis of Caspase-8 and ASC Interactions to Drive Lytic Cell Death, PANoptosis. Mol Biol Evol 2025; 42:msaf096. [PMID: 40277230 PMCID: PMC12066828 DOI: 10.1093/molbev/msaf096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 04/09/2025] [Accepted: 04/15/2025] [Indexed: 04/26/2025] Open
Abstract
Caspases are evolutionarily conserved proteins essential for driving cell death in development and host defense. Caspase-8, a key member of the caspase family, is implicated in nonlytic apoptosis, as well as lytic forms of cell death. Recently, caspase-8 has been identified as an integral component of PANoptosomes, multiprotein complexes formed in response to innate immune sensor activation. Several innate immune sensors can nucleate caspase-8-containing PANoptosome complexes to drive inflammatory lytic cell death, PANoptosis. However, how the evolutionarily conserved and diverse functions of caspase-8 drive PANoptosis remains unclear. To address this, we performed evolutionary, sequence, structural, and functional analyses to decode caspase-8's complex-forming abilities and its interaction with the PANoptosome adaptor ASC. Our study distinguished distinct subgroups within the death domain superfamily based on their evolutionary and functional relationships, identified homotypic traits among subfamily members, and captured key events in caspase evolution. We also identified critical residues defining the heterotypic interaction between caspase-8's death effector domain and ASC's pyrin domain, validated through cross-species analyses, dynamic simulations, and in vitro experiments. Overall, our study elucidated recent evolutionary adaptations of caspase-8 that allowed it to interact with ASC, improving our understanding of critical molecular associations in PANoptosome complex formation and the underlying PANoptotic responses in host defense and inflammation. These findings have implications for understanding mammalian immune responses and developing new therapeutic strategies for inflammatory diseases.
Collapse
Affiliation(s)
- Sivakumar Prasanth Kumar
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Eswar Kumar Nadendla
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - R K Subbarao Malireddi
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Syed Asfarul Haque
- Cryo-Electron Microscopy Center, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Raghvendra Mall
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Andrew F Neuwald
- Institute for Genome Sciences and Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 670 W. Baltimore Street, Baltimore, MD 21201, USA
| | - Thirumala-Devi Kanneganti
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| |
Collapse
|
33
|
Ma W, Wang Q, Guo L, Ju X. The molecular mechanisms, roles, and potential applications of PANoptosis in cancer treatment. Front Immunol 2025; 16:1550800. [PMID: 40364845 PMCID: PMC12069359 DOI: 10.3389/fimmu.2025.1550800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 04/07/2025] [Indexed: 05/15/2025] Open
Abstract
PANoptosis, a newly identified form of programmed cell death regulated by the panoptosome complex, exhibits key characteristics of apoptosis, pyroptosis and necroptosis. It exerts a substantial influence on the initiation and progression of a spectrum of diseases, particularly in cancer, where its impact is increasingly being recognized. PANoptosis is closely related to tumorigenesis, carcinogenesis, metastasis, chemotherapy resistance, as well as the prediction of therapeutic responses and prognosis in cancer patients. In this review, we first review the discovery of PANoptosis and systematically analyze the composition of the panoptosome. Subsequently, we examine the role of PANoptosis in various types of cancer, encompassing its function within the tumor microenvironment, its role in tumor drug resistance, and its predictive role in cancer prognosis. Ultimately, we delve into strategies for targeting PANoptosis in cancer therapy, including targeting various molecules in the PANoptosis pathway, such as ZBP1, RIPK1, RIPK3, Caspases and other novel strategies like nanoinducers and viral vectors. This review aims to provide references and assistance for the research and application of PANoptosis in cancer treatment.
Collapse
Affiliation(s)
- Wenyuan Ma
- Department of Pathology, The People’s Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Danyang Clinical Medical College of Jiangsu University, Danyang, Jiangsu, China
| | - Qiang Wang
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Lanfang Guo
- Department of Clinical Laboratory Medicine, The Fourth People’s Hospital of Jiangsu University, Zhenjiang, Zhenjiang, Jiangsu, China
| | - Xiaoli Ju
- Department of Pathology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| |
Collapse
|
34
|
Min R, Bai Y, Wang NR, Liu X. Gasdermins in pyroptosis, inflammation, and cancer. Trends Mol Med 2025:S1471-4914(25)00090-5. [PMID: 40307076 DOI: 10.1016/j.molmed.2025.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 03/28/2025] [Accepted: 04/04/2025] [Indexed: 05/02/2025]
Abstract
Pyroptosis is a type of programmed inflammatory cell death characterized by balloon-like swelling, membrane rupture, and the release of inflammatory cytokines and danger signals. Pyroptosis is directly triggered by activated gasdermins (GSDMs) which bind to membrane phospholipids, oligomerize, and form pores in cell membranes. GSDM activation is mediated by various effector proteases via cleavage of the linker region or post-translational modification to release the active N-terminal fragment in response to a variety of pathogenic or intrinsic danger signals. GSDM-mediated pyroptosis is involved in the pathogenesis of an array of infectious and inflammatory diseases and cancers. This review discusses recent advances related to the physiological and pathological functions of GSDM-mediated pyroptosis, as well as therapeutic strategies targeting pyroptosis.
Collapse
Affiliation(s)
- Rui Min
- National Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yang Bai
- National Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ning-Rui Wang
- School of Laboratory Medicine, Nanchang Medical College, Nanchang, Jiangxi 330052, China
| | - Xing Liu
- National Key Laboratory of Immune Response and Immunotherapy, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China; Shanghai Academy of Natural Sciences (SANS), Shanghai 200031, China.
| |
Collapse
|
35
|
Jin X, Zhu Y, Xing L, Ding X, Liu Z. PANoptosis: a potential target of atherosclerotic cardiovascular disease. Apoptosis 2025:10.1007/s10495-025-02089-x. [PMID: 40285923 DOI: 10.1007/s10495-025-02089-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/05/2025] [Indexed: 04/29/2025]
Abstract
PANoptosis is a newly discovered cell death pathway triggered by the innate immunizer, which in turn promotes the assembly of the PANoptosome and activates downstream effectors. As a special cell death mode, it is characterized by apoptosis, pyroptosis, and necroptosis at the same time; therefore, it is not feasible to inhibit PANoptosis by suppressing a single cell death pathway. However, active ingredients targeting the PANoptosome can effectively inhibit PANoptosis.Given the importance of cell death in disease, targeting PANoptosis would be an important therapeutic tool. Previous studies have focused more on infectious diseases and cancer, and the role of PANoptosis in the cardiovascular field has not been comprehensively addressed. While ASCVD is the number one killer of cardiovascular diseases, it is important to explore new targets to determine future research directions. Therefore, this review focuses on the assembly of PANoptosome, the molecular mechanism of PANoptosis, and the related mechanisms of PANoptosis leading to ASCVD such as myocardial infarction, ischemic cardiomyopathy and ischemic stroke, in order to provide a new perspective for the prevention and treatment of ASCVD.
Collapse
Affiliation(s)
- Xiao Jin
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
| | - Yanan Zhu
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
| | - Lina Xing
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
| | - Xinyue Ding
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
| | - Zongjun Liu
- Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China.
- , No. 164, Lanxi Road, Putuo District, Shanghai, China.
| |
Collapse
|
36
|
Zhao Y, Liang L, McCaig A, Aujla T, Keshavjee S, Liu M. Activation of PANoptosis and ferroptosis during ex vivo lung perfusion in human lungs. J Heart Lung Transplant 2025:S1053-2498(25)01910-2. [PMID: 40288429 DOI: 10.1016/j.healun.2025.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 11/10/2024] [Accepted: 11/26/2024] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND A recent study demonstrated upregulation of PANoptosis-related genes during reperfusion in human lung transplants. However, the impact of ex vivo lung perfusion (EVLP) on different cell death pathways and their relationship with inflammatory genes and clinical characteristics remains unknown. METHODS We conducted transcriptomic analyses on pre- and post-EVLP biopsies from 49 donation after brain death (DBD) and 39 donation after circulatory death (DCD) lungs. Gene set enrichment analysis (GSEA) and single-sample GSEA were used to assess the enrichment of cell death and inflammatory pathways. We further explored the relationships between these pathways, donor characteristics, and clinical outcomes. RESULTS DBD lungs showed significant enrichment of apoptosis and ferroptosis gene sets compared to DCD lungs. During EVLP, pyroptosis, apoptosis, necroptosis, and ferroptosis gene sets were significantly upregulated and strongly correlated with inflammatory pathways in both DBD and DCD donor lungs. Donor age, sex, and smoking history were associated with specific cell death pathways. In DCD lungs, the expression of ferroptosis-related genes was associated with recipient early outcomes. CONCLUSION The expression of cell death gene sets is donor-type specific. The identification of multiple cell death and inflammatory pathways during EVLP provides potential therapeutic targets to improve donor lung quality and enhance clinical outcomes.
Collapse
Affiliation(s)
- Yajin Zhao
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Lubiao Liang
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Department of Thoracic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, China; Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Abby McCaig
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Tanroop Aujla
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Shaf Keshavjee
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Mingyao Liu
- Latner Thoracic Surgery Research Laboratories, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada; Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada; Department of Surgery, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
37
|
Ye Y, Zhao S, Pang E, Tang Y, Zhu P, Gao W, Diao Q, Yu J, Zeng J, Lan M, Yi J. Indacenodithienothiophene-based A-D-A-type phototheranostics for immuno-phototherapy. J Nanobiotechnology 2025; 23:309. [PMID: 40269932 PMCID: PMC12016427 DOI: 10.1186/s12951-025-03381-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 04/09/2025] [Indexed: 04/25/2025] Open
Abstract
The development of phototherapeutics with high photothermal conversion efficiency (PCE) and strong ability to generate reactive oxygen species under single near-infrared (NIR) laser irradiation for immuno-phototherapy applications remains a significant challenge. Herein, we optimally selected the molecule IT-4 F with an acceptor-donor-acceptor (A-D-A) strucssture to prepare water-dispersible nanoparticles (NPs) by assembly with DSPE-PEG-NH2. Such NPs have NIR absorption and fluorescence peaks at 728 and 817 nm, respectively. They can generate singlet oxygen (1O2) and superoxide anion (O2-·) under laser irradiation, with a 1O2 generation quantum yield of 31.5%. They can also effectively convert photon-energy into heat with a high PCE of 42.8%. The outstanding properties of IT-4 F NPs enable them to be used in NIR fluorescence imaging guided photothermal therapy (PTT), and photodynamic therapy (PDT). Moreover, PDT and PTT triggered immunogenic cell death and PANoptosis in tumor cells, which not only inhibited tumor growth and metastasis in mice model, but also induced a robust immune response, evidenced by increased infiltration of CD8+ T cells, CD4+ T cells, dendritic cells, and a decreased presence of immunosuppressive cells such as myeloid-derived suppressor cells and regulatory T cells. The efficacy of IT-4 F NPs in organoid of human breast cancer was also verified.
Collapse
Affiliation(s)
- Yangtian Ye
- Department of Breast and Thyroid Gland Surgery, Hunan Provincial Maternal And Child Health Care Hospital, Changsha, Hunan, 410008, China
| | - Shaojing Zhao
- College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan, 410083, China
| | - E Pang
- College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan, 410083, China
| | - Yuanyu Tang
- College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan, 410083, China
| | - Pan Zhu
- College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan, 410083, China
| | - Wenjie Gao
- Department of General Surgery, Sir Run Run Hospital of Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Qingxu Diao
- Department of Breast and Thyroid Gland Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, 410005, China
| | - Jie Yu
- Department of Breast and Thyroid Gland Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, 410005, China
| | - Jie Zeng
- Department of Breast and Thyroid Gland Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, 410005, China.
| | - Minhuan Lan
- College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan, 410083, China.
| | - Jianing Yi
- Department of Breast and Thyroid Gland Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, 410005, China.
| |
Collapse
|
38
|
Wu W, Lan W, Jiao X, Wang K, Deng Y, Chen R, Zeng R, Li J. Pyroptosis in sepsis-associated acute kidney injury: mechanisms and therapeutic perspectives. Crit Care 2025; 29:168. [PMID: 40270016 PMCID: PMC12020238 DOI: 10.1186/s13054-025-05329-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 02/21/2025] [Indexed: 04/25/2025] Open
Abstract
Sepsis-associated acute kidney injury (S-AKI) is a severe complication characterized by high morbidity and mortality, driven by multi-organ dysfunction. Recent evidence suggests that pyroptosis, a form of programmed cell death distinct from apoptosis and necrosis, plays a critical role in the pathophysiology of S-AKI. This review examines the mechanisms of pyroptosis, focusing on inflammasome activation (e.g., NLRP3), caspase-mediated processes, and the role of Gasdermin D in renal tubular damage. We also discuss the contributions of inflammatory mediators, oxidative stress, and potential therapeutic strategies targeting pyroptosis, including inflammasome inhibitors, caspase inhibitors, and anti-inflammatory therapies. Lastly, we highlight the clinical implications and challenges in translating these findings into effective treatments, underscoring the need for personalized medicine approaches in managing S-AKI.
Collapse
Affiliation(s)
- Wenyu Wu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, 510405, China
| | - Wanning Lan
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Xin Jiao
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, 510120, China
| | - Kai Wang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Yawen Deng
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, 510120, China
| | - Rui Chen
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Research On Emergency in TCM, Guangzhou, Guangdong, China.
| | - Ruifeng Zeng
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Research On Emergency in TCM, Guangzhou, Guangdong, China.
| | - Jun Li
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| |
Collapse
|
39
|
Zhu J, He T, Huang Z, Yu W, Lu J, Zhang S, Zhang X, Dong H, Xu Y, Wang X, Zhu C. Neutrophil infiltration and microglial shifts in sepsis induced preterm brain injury: pathological insights. Acta Neuropathol Commun 2025; 13:79. [PMID: 40254577 PMCID: PMC12010587 DOI: 10.1186/s40478-025-02002-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 04/07/2025] [Indexed: 04/22/2025] Open
Abstract
Preterm sepsis is a major contributor to brain injury and long-term neurodevelopmental impairments, but its molecular mechanisms remain poorly understood. This study integrated clinical and experimental approaches to investigate the pathological changes linking systemic inflammation to brain injury in preterm infants. Transcriptomic analysis of septic preterm infants' peripheral blood revealed upregulated immune, metabolic, and inflammatory pathways, suggesting a link between systemic and brain inflammation. Using P2 mice, we established a preterm white matter injury model through multiple doses of lipopolysaccharide, observing dose-dependent developmental delays, brain inflammation, and long-term behavioral deficits. Integrative analyses of peripheral blood and brain samples from both mice and preterm infants revealed consistent chemokine alterations and immune cell infiltration across peripheral and central compartments, highlighting the significant involvement of neutrophil extracellular traps in preterm brain injury. Furthermore, microglia exhibited significant transcriptional changes during the acute phase, accompanied by metabolic reprogramming from oxidative phosphorylation to glycolysis, with suggested involvement of Pgk1 and Pgam1. This shift intensified with escalating inflammation, along with PANoptosis-related gene upregulation, ultimately associated with microglial cell death. Collectively, these findings provide pathological insights into the immunometabolic alterations underlying sepsis-induced preterm brain injury and suggest potential targets for future therapeutic interventions to mitigate long-term neurodevelopmental deficits.
Collapse
Affiliation(s)
- Jinjin Zhu
- Henan Key Laboratory of Child Brain Injury and Henan Clinical Research Center for Child Neurological Disorders, Institute of Neuroscience, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Tiantian He
- Henan Key Laboratory of Child Brain Injury and Henan Clinical Research Center for Child Neurological Disorders, Institute of Neuroscience, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Ziwei Huang
- Henan Key Laboratory of Child Brain Injury and Henan Clinical Research Center for Child Neurological Disorders, Institute of Neuroscience, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Wenkai Yu
- Henan Key Laboratory of Child Brain Injury and Henan Clinical Research Center for Child Neurological Disorders, Institute of Neuroscience, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Jinnan Lu
- Henan Key Laboratory of Child Brain Injury and Henan Clinical Research Center for Child Neurological Disorders, Institute of Neuroscience, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Shan Zhang
- Henan Key Laboratory of Child Brain Injury and Henan Clinical Research Center for Child Neurological Disorders, Institute of Neuroscience, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Xiaoli Zhang
- Henan Key Laboratory of Child Brain Injury and Henan Clinical Research Center for Child Neurological Disorders, Institute of Neuroscience, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Huifang Dong
- Henan Key Laboratory of Child Brain Injury and Henan Clinical Research Center for Child Neurological Disorders, Institute of Neuroscience, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yiran Xu
- Henan Key Laboratory of Child Brain Injury and Henan Clinical Research Center for Child Neurological Disorders, Institute of Neuroscience, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Xiaoyang Wang
- Henan Key Laboratory of Child Brain Injury and Henan Clinical Research Center for Child Neurological Disorders, Institute of Neuroscience, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
- Centre of Perinatal Medicine and Health, Institute of Clinical Science, University of Gothenburg, Gothenburg, 40530, Sweden.
| | - Changlian Zhu
- Henan Key Laboratory of Child Brain Injury and Henan Clinical Research Center for Child Neurological Disorders, Institute of Neuroscience, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, 17177, Sweden.
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Goteborg, 40530, Sweden.
| |
Collapse
|
40
|
Song S, Wang J, Ouyang X, Huang R, Wang F, Xie J, Chen Q, Hu D. Therapeutic connections between pyroptosis and paclitaxel in anti-tumor effects: an updated review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04036-8. [PMID: 40257490 DOI: 10.1007/s00210-025-04036-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 03/06/2025] [Indexed: 04/22/2025]
Abstract
As a form of inflammation-associated cell death, pyroptosis has gained widespread attention in recent years. Accumulating evidence indicates that pyroptosis regulates tumor growth and is associated with autoimmune disorders and inflammatory response. Paclitaxel, a traditional Chinese medicine, usually induces death of cancer cells as a chemotherapeutic agent. Previous studies have revealed that paclitaxel can exert an anti-tumor effect through a variety of cell death mechanisms, of which pyroptosis plays a pivotal role in inhibiting tumor growth and enhancing anti-tumor immunity. In this review, we summarize the current advances in therapeutic connections between pyroptosis and paclitaxel in anti-tumor effects.
Collapse
Affiliation(s)
- Shuxin Song
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jingbo Wang
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaohu Ouyang
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Renyin Huang
- Jingshan Union Hospital, Union Hospital, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Fang Wang
- Jingshan Union Hospital, Union Hospital, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Junke Xie
- Jingshan Union Hospital, Union Hospital, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qianyun Chen
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Desheng Hu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- China-Russia Medical Research Center for Stress Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
41
|
He Y, Lu J, Du Y, Zhao L, Gong L, Wu P, Shu Q, Peng H, Wang X. Investigation of PANoptosis pathway in age-related macular degeneration triggered by Aβ1-40. Sci Rep 2025; 15:13514. [PMID: 40251333 PMCID: PMC12008305 DOI: 10.1038/s41598-025-98174-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 04/09/2025] [Indexed: 04/20/2025] Open
Abstract
Our study aimed to identify PANoptosis in Aβ1-40-induced AMD, both in vivo and in vitro, and to determine if AIM2-PANoptosome mediates this process. We used transcriptomics to explore the signaling pathways and target genes linked to PANoptosis within a mouse model of AMD triggered by Aβ1-40. Optical coherence tomography (OCT), hematoxylin and eosin (H&E) staining, and electroretinography (ERG) were employed to assess retinal damage in terms of morphology and function. Morphological changes in ARPE-19 cells were observed using optical microscopy and scanning electron microscopy. Enzyme-linked immunosorbent assay (ELISA) was used to detect the levels of cytokines in cell supernatants, mouse orbital serum, and human plasma to evaluate the severity of inflammation. CO-immunoprecipitation(CoIP) and molecular docking were performed to assess the impact and expression of proteins associated with the AIM2-PANoptosome. Quantitative polymerase chain reaction (qPCR), Western blot (WB), immunofluorescence, and apoptosis detection kits were used to evaluate the expression levels of genes and proteins related to PANoptosis-like cell death. Our results showed that the Aβ1-40-induced AMD model had increased expression of apoptosis, necroptosis, and pyroptosis pathways, and AIM2-PANoptosome components. CoIP and docking confirmed increased AIM2, ZBP1, and PYRIN levels under Aβ1-40 treatment. WB and immunofluorescence showed upregulation of PANoptosis-related proteins. Inhibitors reduced Aβ-induced protein expression. ELISA showed increased inflammatory cytokines. Apoptosis assays and microscopy revealed Aβ1-40-induced ARPE-19 cell loss and morphological changes. In conclusion, the Aβ1-40-induced AMD model displayed PANoptosis-like cell death, offering insights into disease pathogenesis.
Collapse
Affiliation(s)
- Yuxia He
- Department of Ophthalmology, Chongqing Key Laboratory for the Prevention and Treatment of Major Blinding Eye Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Guiyang Aier Eye Hospital, Guiyang, Guizhou Province, China
| | - Jing Lu
- Department of Ophthalmology, Chongqing Key Laboratory for the Prevention and Treatment of Major Blinding Eye Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yong Du
- Department of Ophthalmology, Chongqing Key Laboratory for the Prevention and Treatment of Major Blinding Eye Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Long Zhao
- Department of Ophthalmology, Chongqing Key Laboratory for the Prevention and Treatment of Major Blinding Eye Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Lili Gong
- Guiyang Aier Eye Hospital, Guiyang, Guizhou Province, China
| | - Ping Wu
- Department of Ophthalmology, Chongqing Key Laboratory for the Prevention and Treatment of Major Blinding Eye Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Qinxin Shu
- Department of Ophthalmology, Chongqing Key Laboratory for the Prevention and Treatment of Major Blinding Eye Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Hui Peng
- Department of Ophthalmology, Chongqing Key Laboratory for the Prevention and Treatment of Major Blinding Eye Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Xing Wang
- Department of Ophthalmology, Chongqing Key Laboratory for the Prevention and Treatment of Major Blinding Eye Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
42
|
Sharma BR, Choudhury SM, Abdelaal HM, Wang Y, Kanneganti TD. Innate immune sensor NLRP3 drives PANoptosome formation and PANoptosis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkaf042. [PMID: 40249072 DOI: 10.1093/jimmun/vkaf042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 01/30/2025] [Indexed: 04/19/2025]
Abstract
Inflammasomes are multiprotein innate immune complexes formed in response to infections, tissue damage, or cellular stress that promote the maturation and release of IL-1β/IL-18 and are implicated in lytic cell death. The NLRP3 inflammasome is canonically activated by an initial priming event followed by an activation stimulus, leading to rapid cell death that occurs through caspase-1 (CASP1) and gasdermin D (GSDMD) activation, called pyroptosis. CASP1- and GSDMD-deficient cells are protected from the rapid LPS plus ATP-induced pyroptosis. However, innate immune responses physiologically occur over time, extending beyond minutes to hours and days. Therefore, in this study, we assessed lytic cell death beyond the early timepoints. While cells lacking the innate immune sensor NLRP3 were protected from cell death induced by the canonical NLRP3 trigger, LPS priming and ATP stimulation (LPS plus ATP), for extended time, CASP1- and GSDMD-deficient cells started to lyse in a time-dependent manner after 2 h. Nevertheless, robust IL-1β and IL-18 release was still dependent on CASP1 activation. These data suggested that NLRP3 engages an additional innate immune, lytic cell death pathway. Indeed, LPS plus ATP induced the activation of caspases and RIPKs associated with PANoptosis in WT cells, and cells deficient in PANoptosis machinery were protected from cell death for extended times. A PANoptosome complex containing NLRP3, ASC, CASP8, and RIPK3 was observed by microscopy in WT, as well as CASP1- or GSDMD-deficient, cells by 30 min post-stimulation. Overall, these findings highlight the central role of NLRP3 as a PANoptosome sensor. Given the physiological role of innate immune cell death, PANoptosis, in health and disease, our study emphasizes the importance of a comprehensive understanding of PANoptosomes, and their components, as therapeutic targets.
Collapse
Affiliation(s)
- Bhesh Raj Sharma
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Sk Mohiuddin Choudhury
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Hadia M Abdelaal
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Yaqiu Wang
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, United States
| | | |
Collapse
|
43
|
Chen W, Jiang Y, Zeng J, Liu D, Feng X, Cheng Y, Lu D, Sun Y, Zhu Q, Zhang X, Wang Q. FDX1 promotes elesclomol-induced PANoptosis in diffuse large B-cell lymphoma via activating IRF3/IFN-β signaling. Oncogene 2025:10.1038/s41388-025-03366-4. [PMID: 40240522 DOI: 10.1038/s41388-025-03366-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 03/02/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025]
Abstract
Diffuse large B-cell lymphoma (DLBCL) remains a major clinical challenge and requires the development of new therapeutic approaches. The identification of cuproptosis, a newly defined form of copper-induced cell death, has provided innovative insights for cancer therapy. Here, we report that loss of the mitochondrial matrix reductase FDX1 in DLBCL cells impairs the antitumor effect of elesclomol (ES), which performs its function by transporting excess copper into cells. Overexpressing (OE) FDX1 significantly sensitized DLBCL cells to ES-induced cell death in vitro and enhanced the anticancer activity of ES in vivo. Furthermore, treatment with ES in FDX1-high expression patient-derived xenograft (PDX) showed a significantly greater inhibitory effect than in FDX1-low expression PDX. Mechanistically, FDX1 promotes the induction of IFN-β-dependent PANoptosis by increasing IRF3 phosphorylation in DLBCL cells upon ES treatment. Consistent with this finding, patient cohort analysis revealed that FDX1 expression correlated positively with enhanced IRF3 phosphorylation. Together, our findings are the first to identify the central role of FDX1 in synergizing with ES to activate IFN-β signaling and induce PANoptosis. This study enables us to re-explore the clinical anticancer potential of ES as a novel therapeutic strategy for DLBCL.
Collapse
Affiliation(s)
- Weifeng Chen
- Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, 511518, China
- Affiliated Cancer Hospital/Institute and GMU-GIBH Joint School of Life Sciences of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou, 511495, China
| | - Yuhang Jiang
- Department of Orthopedics, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518033, China
| | - Jun Zeng
- Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, 511518, China
| | - Dandan Liu
- Affiliated Cancer Hospital/Institute and GMU-GIBH Joint School of Life Sciences of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou, 511495, China
| | - Xiaoting Feng
- Affiliated Cancer Hospital/Institute and GMU-GIBH Joint School of Life Sciences of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou, 511495, China
| | - Yiqiu Cheng
- Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, 511518, China
| | - Di Lu
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Yangbai Sun
- Department of Musculoskeletal Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
| | - Qinyuan Zhu
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, 311599, China.
| | - Xiaoren Zhang
- Affiliated Cancer Hospital/Institute and GMU-GIBH Joint School of Life Sciences of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangzhou, 511495, China.
| | - Qi Wang
- Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, 511518, China.
| |
Collapse
|
44
|
Li J, Jia YC, Lu J, Zhang H, Wang Z, Xie X, Cao F, Li F. Inhibition of Zbp1-PANoptosome-mediated PANoptosis effectively attenuates acute pancreatitis. Cell Death Discov 2025; 11:180. [PMID: 40240343 PMCID: PMC12003674 DOI: 10.1038/s41420-025-02451-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 03/21/2025] [Accepted: 03/27/2025] [Indexed: 04/18/2025] Open
Abstract
Early acute pancreatitis is an acute inflammatory disease that involves multiple modes of cell death, including apoptosis, necrotic apoptosis, and pyroptosis in its disease process. PANoptosis, a type of cell death that includes pyroptosis, apoptosis, and necroptosis, has had an important role in a variety of infectious and inflammatory diseases in recent years. To judge the relationship between PANoptosis and AP, we first analyzed the data from pancreatic transcriptome data by bioinformatics techniques, and we found the enrichment of PANoptosis pathway in AP. Next, we screened the genes and identified differentially expressed genes (DEGs) associated with AP and PANoptosis. Finally, we found that Zbp1 may have a major role in the process of PANoptosis. For this purpose, we constructed AP models in mice and in vitro cell line 266-6 and intervened by inhibiting Zbp1. The final results showed that the PANoptosis in mice was significantly suppressed after inhibition of Zbp1. In conclusion, inflammatory injury in AP can be significantly improved by inhibiting Zbp1- PANoptosome-mediated PANoptosis.
Collapse
Affiliation(s)
- Jie Li
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Clinical Center for Acute Pancreatitis, Capital Medical University, Beijing, China
| | - Yu-Chen Jia
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Clinical Center for Acute Pancreatitis, Capital Medical University, Beijing, China
| | - Jiongdi Lu
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Clinical Center for Acute Pancreatitis, Capital Medical University, Beijing, China
| | - Haoyu Zhang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Clinical Center for Acute Pancreatitis, Capital Medical University, Beijing, China
| | - Zheng Wang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Clinical Center for Acute Pancreatitis, Capital Medical University, Beijing, China
| | - Xiaozhou Xie
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Clinical Center for Acute Pancreatitis, Capital Medical University, Beijing, China
| | - Feng Cao
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Clinical Center for Acute Pancreatitis, Capital Medical University, Beijing, China
| | - Fei Li
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China.
- Clinical Center for Acute Pancreatitis, Capital Medical University, Beijing, China.
| |
Collapse
|
45
|
Yang Y, Chen X, Deng L, Huang Y, Mo Y, Ye J, Liang R, Qin Y, Zhang Q, Wang S. Arsenic exposure provoked prostatic PANoptosis by inducing mitochondrial dysfunction in mice and WPMY-1 cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 295:118139. [PMID: 40185034 DOI: 10.1016/j.ecoenv.2025.118139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 03/27/2025] [Accepted: 03/31/2025] [Indexed: 04/07/2025]
Abstract
Inorganic arsenic, a widespread environmental toxicant, significantly contributes to prostate injury. However, the exact cellular mechanisms remain unclear. This study explored the involvement of pyroptosis, apoptosis, and necroptosis (PANoptosis), and their interconnections in arsenic-induced prostate injury. Herein, by employing in vitro (WPMY-1 cells exposed to arsenic for 48 h with or without reactive oxygen species (ROS) and mitochondrial ROS scavenger treatments) and in vivo (C57BL/6 mice were orally gavaged with arsenic and/or N-acetylcysteine for 90 consecutive days) models of arsenic-induced prostate injury and intervention, we demonstrated that sodium arsenite (NaAsO2) triggered mitochondrial damage-activated PANoptosis via the Bax/Bcl-xL/caspase-3/Gasdermin E (GSDME) pathway and the Z-DNA binding protein 1/receptor-interacting protein kinases 1 (RIPK1)/RIPK3/mixed lineage kinase domain-like protein (MLKL) signaling pathway. Notably, treatment with NaAsO2, GSDME, or MLKL knockdown in WPMY-1 cells increased the phenotype of PANoptosis. Mechanistically, the GSDME-N, GSDMD-N, p-MLKL, and cleaved caspase-3 protein levels were increased (1.4-, 2.67-, 3.51-, and 2.16-fold, respectively) in NaAsO2-treated GSDME knockdown WPMY-1 cells, whereas GSDME-N and cleaved caspase-3 protein levels were increased (1.30- and 1.21-fold, respectively) in NaAsO2-treated MLKL knockdown WPMY-1 cells. Our study highlights the crucial role of mitochondrial dysfunction in the initiation of PANoptosis during arsenic-induced prostate injury. Furthermore, we provide novel insights into the connections between apoptosis, pyroptosis, and necroptosis, indicating that GSDME and MLKL proteins may act as crucial regulators and potential therapeutic targets for arsenic-induced PANoptosis.
Collapse
Affiliation(s)
- Yiping Yang
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Xianglan Chen
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Longxin Deng
- Department of Urology, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Yurun Huang
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Yingxi Mo
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Jiazhou Ye
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Rong Liang
- Department of Digestive Oncology, Guangxi Medical University Cancer Hospital, Nanning 530021, China
| | - Yaxin Qin
- The Second Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Qingyun Zhang
- Department of Urology, Guangxi Medical University Cancer Hospital, Nanning 530021, China.
| | - Shan Wang
- Department of Experimental Research, Guangxi Medical University Cancer Hospital, Nanning 530021, China.
| |
Collapse
|
46
|
Cui J, Wu D, Lv D. Predicting survival and immune status of breast cancer patients based on prognostic features related to PANoptosis. Discov Oncol 2025; 16:527. [PMID: 40232576 PMCID: PMC11999922 DOI: 10.1007/s12672-025-02209-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 03/21/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND Breast cancer (BRCA) is a prevalent female malignancy. PANoptosis, integrating diverse cell death traits, is pivotal in BRCA, thus necessitating deeper study. METHODS Data from Gene Expression Omnibus (GEO, GSE180286 and GSE20685) and The Cancer Genome Atlas (TCGA) were analyzed. Weighted gene co-expression network analysis (WGCNA) identified PANoptosis-related genes in BRCA patients from TCGA. Further refinement of these module genes was conducted through univariate Cox regression, LASSO regression (glmnet package), and stepwise multivariate regression analysis to derive the final biomarkers. Based on these biomarkers, a risk model was established, and in-vitro experiments (wound healing assay, Transwell assay, and qRT-PCR) were carried out to validate the accuracy of these biomarkers. The MCPcounter package and the oncoPredict package were used to assess immune cell infiltration and sensitivity to drugs in BRCA patients, respectively. RESULTS This study identified 8 biomarkers (ACY3, CD83, CXCL13, KLHDC7B, NR1H3, SMCO4, TRPM2, and UPP1) and established a risk model. In-vitro experiments revealed significant differences in biomarker expression between BRCA cells and the control group, with TRPM2 knockdown inhibiting BRCA cell migration and invasion. Enrichment analysis showed metabolic pathways were activated in high-risk group. Additionally, immune analysis showed lower immune cell enrichment and significant enrichment of fibroblasts in the high-risk group. Drug sensitivity analysis linked 13 drugs to RiskScore. Finally, single-cell analysis identified six cell types (including cancer stem cells, fibroblasts, T-cells, macrophages, B/Plasma cells, and endothelial cells) for BRCA and found that macrophages had higher PANoptosis activity. CONCLUSION The current research introduces a novel model for BRCA prognosis analysis but also provides a fresh perspective on BRCA treatment strategies.
Collapse
Affiliation(s)
- Juanjuan Cui
- Department of Oncology, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, 266071, China
| | - Dapeng Wu
- Department of Oncology, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, 266071, China
| | - Da Lv
- Department of Oncology, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, 266071, China.
| |
Collapse
|
47
|
Wan C, Wu Q, Wang Y, Sun Y, Ji T, Gu Y, Wang L, Chen Q, Yang Z, Wang Y, Wang B, Zhong W. Machine learning-based characterization of PANoptosis-related biomarkers and immune infiltration in ulcerative colitis: A comprehensive bioinformatics analysis and experimental validation. Int Immunopharmacol 2025; 151:114298. [PMID: 39986196 DOI: 10.1016/j.intimp.2025.114298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 11/25/2024] [Accepted: 02/11/2025] [Indexed: 02/24/2025]
Abstract
Ulcerative colitis (UC) is a heterogeneous autoimmune condition. PANoptosis, a new form of programmed cell death, plays a role in inflammatory diseases. This study aimed to identify differentially expressed PANoptosis-related genes (PRGs) involved in immune dysregulation in UC. Three key PRGs-BIRC3, MAGED1, and PSME2 were found using weighted gene co-expression network analysis (WGCNA) and machine learning. Immune infiltration analysis revealed that these key PRGs were associated with neutrophils, CD8+ T cells, activated CD4 T cells, and NK cells. Moreover, these key PRGs were significantly enriched in pathways related to inflammatory bowel disease, the IL-17 signaling pathway, and NOD-like receptor signaling pathway. The expression levels of the key PRGs were validated in various datasets, animal models, and UC intestinal tissue samples. Our findings confirmed the involvement of PANoptosis in UC and predict hub genes and immune characteristics, providing new insights for further investigations into UC pathogenic mechanisms and therapeutic strategies.
Collapse
Affiliation(s)
- Changshan Wan
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin 300052, China
| | - Qiuyan Wu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin 300052, China
| | - Yali Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin 300052, China
| | - Yan Sun
- Department of Obstetrics and Gynecology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Tao Ji
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin 300052, China; Department of Digestive Gastroenterology and Hepatology, Linyi People's Hospital, Shandong 276000, China
| | - Yu Gu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin 300052, China
| | - Liwei Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin 300052, China
| | - Qiuyu Chen
- Department of Gastroenterology, Tianjin First Central Hospital of Tianjin Medical University, Tianjin 300192, China
| | - Zhen Yang
- Department of Clinical Laboratory, Tianjin Cancer Institute of Integrative Traditional Chinese and Western Medicine, Tianjin Union Medical Center of Nankai University, China.
| | - Yao Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine,Harbin 150040, China.
| | - Bangmao Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin 300052, China.
| | - Weilong Zhong
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin 300052, China.
| |
Collapse
|
48
|
Griffioen AW, Nowak-Sliwinska P. A cellular danse macabre: the choreography of programmed cell death. Apoptosis 2025; 30:507-511. [PMID: 39924582 DOI: 10.1007/s10495-025-02076-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/02/2025] [Indexed: 02/11/2025]
Abstract
Research on cell death is getting diverse. Apoptosis and other forms of programmed cell death play a critical role in maintaining cellular homeostasis and defending an organism from infections, cancer, and degenerative diseases. Apoptosis, a well-known form of programmed cell death, involves non-inflammatory and orderly organized dismantling of a cell. Different pathways and mechanisms have emerged that challenge the traditional apoptosis-centric view, such as necroptosis, panoptosis, pyroptosis, paraptosis, ferroptosis and autophagic cell death. This editorial aims to highlight some of these emerging pathways, expanding our understanding of cellular death and its implications in health and disease. Over the years, Apoptosis has been at the forefront of publishing discoveries on these diverse and impactful processes.
Collapse
Affiliation(s)
- Arjan W Griffioen
- Angiogenesis Laboratory, Department of Medical Oncology, Amsterdam UMC, Cancer Center Amsterdam, Amsterdam, The Netherlands.
| | | |
Collapse
|
49
|
Xiong W, Li J, Tian A, Mao X. Unravelling the Role of PANoptosis in Liver Diseases: Mechanisms and Therapeutic Implications. Liver Int 2025; 45:e70000. [PMID: 40116786 DOI: 10.1111/liv.70000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 12/28/2024] [Accepted: 01/08/2025] [Indexed: 03/23/2025]
Abstract
PANoptosis is a multimodal form of cell death that involves inflammatory, apoptotic, and necroptotic pathways, playing a key role in the development of liver diseases. This article first outlines the definition and characteristics of PANoptosis, and then explores its mechanisms of action in different types of liver diseases, including acute liver injury, liver failure, metabolic dysfunction-associated fatty liver disease, and hepatocellular carcinoma. Furthermore, this article analyses the molecular regulatory network of PANoptosis and potential therapeutic targets. Finally, this article summarises the current research on PANoptosis in liver diseases and future research directions, and it reviews the role of the emerging cell death mechanism of PANoptosis in liver diseases.
Collapse
Affiliation(s)
- Wanyuan Xiong
- The First Clinical Medical College of Lanzhou University, Lanzhou University, Lanzhou, Gansu, China
- Department of Infectious Disease, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Junfeng Li
- The First Clinical Medical College of Lanzhou University, Lanzhou University, Lanzhou, Gansu, China
- Department of Infectious Disease, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
- Department of Liver Disease, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Aiping Tian
- Department of Infectious Disease, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Xiaorong Mao
- Department of Infectious Disease, The First Hospital of Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
50
|
Hou Y, Feng Q, Wei C, Cao F, Liu D, Pan S, Shi Y, Liu Z, Liu F. Emerging role of PANoptosis in kidney diseases: molecular mechanisms and therapeutic opportunities. Apoptosis 2025; 30:579-596. [PMID: 39833634 DOI: 10.1007/s10495-024-02072-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/26/2024] [Indexed: 01/22/2025]
Abstract
Kidney diseases represent a significant global public health challenge, characterized by complex pathogenesis, high incidence, low awareness, insufficient early screening, and substantial treatment disparities. Effective therapeutic options remain lacking. Programmed cell death (PCD), including apoptosis, pyroptosis, and necroptosis, play pivotal roles in the pathogenesis of various kidney diseases. In 2019, PANoptosis, a novel form of inflammatory cell death, was introduced, providing new insights into innate immunity and PCD research. Although research on PANoptosis in kidney diseases is still limited, identifying key molecules within PANoptosomes and understanding their regulatory roles is critical for disease prevention and management. This review summarizes the various forms of PCD implicated in kidney diseases, along with PANoptosomes activated by Z-DNA binding protein 1 (ZBP1), absent in melanoma 2 (AIM2), receptor-interacting protein kinase 1 (RIPK1), NOD-like receptor family CARD domain containing 12 (NLRP12), and NOD-like receptor family member C5 (NLRC5). It also reviews the advancements in PANoptosis research in the field of kidney diseases, particularly in renal tumors and acute kidney injuries (AKI). The goal is to establish a foundation for future research into the role of PANoptosis in kidney diseases.
Collapse
Affiliation(s)
- Yi Hou
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, P. R. China
| | - Qi Feng
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, P. R. China
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, P. R. China
| | - Cien Wei
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, P. R. China
| | - Fengyu Cao
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, P. R. China
| | - Dongwei Liu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, P. R. China
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, P. R. China
| | - Shaokang Pan
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, P. R. China
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, P. R. China
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, P. R. China
| | - Yan Shi
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, P. R. China
| | - Zhangsuo Liu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China.
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China.
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, P. R. China.
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, P. R. China.
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, P. R. China.
| | - Fengxun Liu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China.
- Traditional Chinese Medicine Integrated Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, P. R. China.
- Research Institute of Nephrology, Zhengzhou University, Zhengzhou, 450052, P. R. China.
- Henan Province Research Center for Kidney Disease, Zhengzhou, 450052, P. R. China.
- Key Laboratory of Precision Diagnosis and Treatment for Chronic Kidney Disease in Henan Province, Zhengzhou, 450052, P. R. China.
| |
Collapse
|