1
|
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] [MESH Headings] [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
|
2
|
Zhao Q, Zhang X, Zhang J, Zhang Y, Jia L, Guo S, Zhang M, Wang H, Wang Y, Guan Y, Zhang Y, Miao S, Zhu JX, Ma H. Reduction of D2 receptors on microglia leads to ZBP1-mediated PANoptosis of mPFC in Parkinson's disease depression mice. Int Immunopharmacol 2025; 158:114809. [PMID: 40367691 DOI: 10.1016/j.intimp.2025.114809] [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: 03/18/2025] [Revised: 04/28/2025] [Accepted: 05/04/2025] [Indexed: 05/16/2025]
Abstract
Parkinson's disease depression (PDD) is a common non-motor symptom of Parkinson's disease (PD), characterized by complex neurobiological mechanisms that remain poorly understood. This study identifies ZBP1-mediated PANoptosis as a critical mechanism linking neuroinflammation, neuronal loss, and depressive behaviors in PDD. Using a 6-hydroxydopamine (6-OHDA)-induced PDD mouse model, we observed significant reductions in dopaminergic projections from the substantia nigra (SN) to the medial prefrontal cortex (mPFC), accompanied by neuronal loss and depressive-like behaviors. Microglial activation, driven by DRD2 downregulation, was found to impair mPFC neuronal function, as evidenced by altered local field potentials and reduced gamma, beta, and theta oscillations. Furthermore, ZBP1 expression was significantly upregulated in the mPFC of PDD mice, where it colocalized with CaMKII-positive neurons and facilitated the formation of PANoptosomes, a multimeric complex driving pyroptosis, apoptosis, and necroptosis. Knockdown of ZBP1 in the mPFC effectively suppressed PANoptosome formation, reduced neuronal injury, restored local field potentials, and alleviated depressive-like behaviors. These findings highlight ZBP1-mediated PANoptosis as a key pathological mechanism in PDD and suggest that targeting ZBP1 may represent a promising therapeutic strategy for mitigating neuronal loss and depressive symptoms in PDD.
Collapse
Affiliation(s)
- Qiyue Zhao
- Department of Physiology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xinyao Zhang
- Department of Physiology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jing Zhang
- Department of Physiology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Ying Zhang
- Department of Physiology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Lu Jia
- Department of Physiology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Sijia Guo
- Department of Physiology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Mengqing Zhang
- Department of Physiology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Haoran Wang
- Department of Physiology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yuling Wang
- Department of Neurology, the Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yue Guan
- Department of Physiology, Hebei Medical University, Shijiazhuang, Hebei, China; Key Laboratory of Neurophysiology of Hebei Province, Shijiazhuang, China
| | - Yi Zhang
- Department of Physiology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Suibing Miao
- Institute of Reproductive Medicine of Shijiazhuang, The Fourth Hospital of Shijiazhuang, Gynecology and Obstetrics Hospital Affiliated to Hebei Medical University, Shijiazhuang, China
| | - Jin-Xia Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Science, Capital Medical University, No. 10 Xitoutiao, You An Men, Beijing, China
| | - Huijie Ma
- Department of Physiology, Hebei Medical University, Shijiazhuang, Hebei, China; Key Laboratory of Neurophysiology of Hebei Province, Shijiazhuang, China; The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang, Hebei, China.
| |
Collapse
|
3
|
Dai M, Dai X, Liang Y, Li X, Huang H, Zhao W. Keratinocyte necroptosis promotes the progression of radiation-induced oral mucositis. BMC Oral Health 2025; 25:941. [PMID: 40483488 PMCID: PMC12145616 DOI: 10.1186/s12903-025-06278-7] [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: 01/05/2025] [Accepted: 05/26/2025] [Indexed: 06/11/2025] Open
Abstract
IMPORTANCE Radiation-induced oral mucositis (RIOM) is a prevalent complication arising from radiation therapy for tumors or combined radiotherapy, but the therapeutic options available remain limited. Understanding its underlying mechanisms is crucial for developing effective interventions. OBJECTIVES To investigate whether keratinocyte necroptosis contributes to RIOM pathogenesis and evaluate the effects of RIPK3/MLKL inhibition. METHODS A mouse model of RIOM was established with varying irradiation doses. Tongue tissues were analyzed via histological staining, immunohistochemistry, and Western blot. In vitro, keratinocytes were irradiated and treated with RIPK3 or MLKL inhibitors. Subsequently, cell viability, necroptosis, and inflammatory cytokine expression were assessed using CCK-8, LDH release, Western blot, flow cytometry and RT-qPCR. RESULTS In irradiated mouse tongues, p-RIPK3/RIPK3 and p-MLKL/MLKL ratios were significantly elevated (P < 0.01), accompanied by heightened expression levels of IL-1β and IL-6. Similar findings were observed in keratinocytes, which, after 12 Gy irradiation for 2.5 days, reduced cell viability (P < 0.001), enhanced necroptotic marker expression (P < 0.001), and increased inflammatory cytokine levels (P < 0.001). Furthermore, treatment with RIPK3 inhibitor GSK'872 or MLKL inhibitor GW806742X significantly reduced irradiation-induced keratinocyte cell death (P < 0.001), LDH release (P < 0.001) and the expression of inflammatory cytokines (P < 0.01). CONCLUSIONS This study provides evidence that RIPK3/MLKL-mediated necroptosis in keratinocytes contributes to the pathogenesis of RIOM. Inhibiting this pathway reduces cell death and inflammation, suggesting a promising therapeutic target for the treatment of RIOM.
Collapse
Affiliation(s)
- Manqiong Dai
- Department of Stomatology, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, Guangzhou, 510515, China
| | - Xingzhu Dai
- Department of Stomatology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yuee Liang
- Department of Stomatology, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, Guangzhou, 510515, China
| | - Xiaoyu Li
- Department of Stomatology, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, Guangzhou, 510515, China
| | - Huacong Huang
- Department of Stomatology, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, Guangzhou, 510515, China
| | - Wanghong Zhao
- Department of Stomatology, Nanfang Hospital, Southern Medical University, No. 1838, Guangzhou Avenue North, Guangzhou, 510515, China.
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
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
|
7
|
Salim Abed H, Oghenemaro EF, Kubaev A, Jeddoa ZMA, S R, Sharma S, Vashishth R, Jabir MS, Jawad SF, Zwamel AH. Non-coding RNAs as a Critical Player in the Regulation of Inflammasome in Inflammatory Bowel Diseases; Emphasize on lncRNAs. Cell Biochem Biophys 2025; 83:1359-1374. [PMID: 39424765 DOI: 10.1007/s12013-024-01585-2] [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: 09/28/2024] [Indexed: 10/21/2024]
Abstract
Inflammatory bowel disease (IBD) is an idiopathic disease caused by a dysregulated immune response to host intestinal microflora. A hyperactive inflammatory and immunological response in the gut has been shown to be one of the disease's long-term causes despite the complexity of the clinical pathology of IBD. The innate immune system activator known as human gut inflammasome is thought to be a significant underlying cause of pathology and is closely linked to the development of IBD. It is essential to comprehend the function of inflammasome activation in IBD to treat it effectively. Systemic inflammasome regulation may be a proper therapeutic and clinical strategy to manage IBD symptoms since inflammasomes may have a significant function in IBD. Non-coding RNAs (ncRNAs) are a type of RNA transcript that is incapable of encoding proteins or peptides. In IBD, inflammation develops and worsens as a result of its imbalance. Culminating evidence has been shown that ncRNAs, and particularly long non-coding RNAs (lncRNAs), may play a role in the regulation of NLR family pyrin domain containing 3 (NLRP3) inflammasome activation in IBD. The relationship between IBD and the gut inflammasome, as well as current developments in IBD research and treatment approaches, have been the main topics of this review. We have covered inflammasomes and their constituents, results from in vivo research, inflammasome inhibitors, and advancements in inflammasome-targeted therapeutics for IBD.
Collapse
Affiliation(s)
- Hussein Salim Abed
- Department of Medical Laboratory Techniques, Al-Maarif University College, Al-Anbar, Ramadi, Iraq
| | - Enwa Felix Oghenemaro
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Delta State University, Abraka, Delta State, Nigeria.
| | - Aziz Kubaev
- Department of Maxillofacial Surgery, Samarkand State Medical University, 18 Amir Temur Street, Samarkand, 140100, Uzbekistan
| | | | - RenukaJyothi S
- Department of Biotechnology and Genetics, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Shilpa Sharma
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, 140307, India
| | - Raghav Vashishth
- Department of Surgery, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| | - Majid S Jabir
- Department of Applied Sciences, University of Technology, Karbala, Iraq
| | - Sabrean Farhan Jawad
- Department of Biochemistry, College of Science, Al-Mustaqbal University, 51001, Babil, Iraq
| | - Ahmed Hussein Zwamel
- Medical laboratory technique college, the Islamic University, Najaf, Iraq
- Medical laboratory technique college, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Medical laboratory technique college, the Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
8
|
Wang P, Zhang C, Wu P, Zhao Z, Sun N, Xue Q, Gao S, He J. Cell Death and Senescence-Based Molecular Classification and an Individualized Prediction Model for Lung Adenocarcinoma. MedComm (Beijing) 2025; 6:e70237. [PMID: 40443719 PMCID: PMC12122187 DOI: 10.1002/mco2.70237] [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: 01/12/2024] [Revised: 03/31/2025] [Accepted: 04/10/2025] [Indexed: 06/02/2025] Open
Abstract
The exploration of cell death and cellular senescence (CDS) in cancer has been an area of interest, yet a systematic evaluation of CDS features and their interactions in lung adenocarcinoma (LUAD) to understand tumor heterogeneity, tumor microenvironment (TME) characteristics, and patient clinical outcomes is previously uncharted. Our study characterized the activities and interconnections of 21 CDS features in 1788 LUAD cases across 15 cohorts, employing unsupervised clustering to categorize patients into three CDS subtypes with distinct TME profiles. The CDS index (CDSI), derived from principal component analysis, was developed to assess individual tumor CDS regulation patterns. Twelve CDSI core genes, enriched in proliferating T cells within the TME as per single-cell analysis, were identified and their functional roles and prognostic significance were validated. High CDSI correlated with improved overall survival in discovery cohort, four independent validation cohorts, and subgroup analysis. CDSI-low patients exhibited a favorable clinical response to immunotherapy and potential sensitivity to mitosis pathway drugs, while CDSI-high patients might benefit from drugs targeting ERK/MAPK and MDM2-p53 pathways. The clinical utility of CDSI was further validated using 9185 pan-cancer samples, demonstrating the broad relevance of our prediction model across various cancer types and its potential clinical implications for cancer management.
Collapse
Affiliation(s)
- Pan Wang
- Department of Thoracic SurgeryNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Chaoqi Zhang
- Department of Thoracic SurgeryNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Peng Wu
- Department of Thoracic SurgeryNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Zhihong Zhao
- Department of Thoracic SurgeryNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Nan Sun
- Department of Thoracic SurgeryNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Qi Xue
- Department of Thoracic SurgeryNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Shugeng Gao
- Department of Thoracic SurgeryNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Jie He
- Department of Thoracic SurgeryNational Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
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
|
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
|
11
|
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
|
12
|
Műzes G, Sipos F. PANoptosis as a Two-Edged Sword in Colorectal Cancer: A Pathogenic Mechanism and Therapeutic Opportunity. Cells 2025; 14:730. [PMID: 40422233 DOI: 10.3390/cells14100730] [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: 04/06/2025] [Revised: 05/14/2025] [Accepted: 05/16/2025] [Indexed: 05/28/2025] Open
Abstract
The examination of PANoptosis in colorectal cancer is particularly important, as many tumor cells can evade apoptotic cell death while continuing to proliferate through inflammatory mediators and creating an immunosuppressive environment. The PANoptosome functions as a regulatory complex that unites proteins governing pyroptotic, apoptotic, and necroptotic pathways, rather than allowing distinct death pathways to compete. The expression and functional status of key molecules within the PANoptosome, such as ZBP1, RIPK1, RIPK3, CASP8, and ASC, may influence tumor viability and immune detection. The tumorigenic impact of PANoptosis is complex and predominantly manifests through chronic inflammation, immune response modulation, and changes in the tumor microenvironment. PANoptosis also aids in the defense against colon cancer by directly eradicating tumor cells and modifying the cellular environment. The expression profile of PANoptosis components may possess prognostic and predictive significance. The therapeutic ramifications of PANoptosis in colorectal cancer are now being investigated through many avenues. It provides an opportunity to develop targeted therapeutic techniques. In contrast, it may also be pertinent in conjunction with immunotherapy, as PANoptosis signifies an immunogenic type of cell death and may consequently enhance the anti-tumor immune response. A thorough comprehension of how these parameters influence PANoptosis is crucial for practical implementation.
Collapse
Affiliation(s)
- Györgyi Műzes
- Immunology Division, Department of Internal Medicine and Hematology, Semmelweis University, 1088 Budapest, Hungary
| | - Ferenc Sipos
- Immunology Division, Department of Internal Medicine and Hematology, Semmelweis University, 1088 Budapest, Hungary
| |
Collapse
|
13
|
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
|
14
|
Lin Z, Wang J, Luo H, Huang L, Pan Z, Yang S, Zhong C, Shan NC, Ye Z, Tan H, Yang X, Zhang B, Huang C, Zhang H. Changdiqing decoction (CDQD) ameliorates colitis via suppressing inflammatory macrophage activation and modulating gut microbiota. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 143:156856. [PMID: 40412060 DOI: 10.1016/j.phymed.2025.156856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 05/11/2025] [Accepted: 05/13/2025] [Indexed: 05/27/2025]
Abstract
BACKGROUND Ulcerative colitis (UC) is a non-specific inflammatory bowel disease. Unlike any single form of cell death reported previously, macrophage PANoptosis, a unique programmed cell death characterized by inflammation and necrosis, plays a crucial role in the pathogenesis of colitis. Changdiqing Decoction (CDQD), an empirical hospital prescription enema, has been used to treat UC for decades. This study aimed to investigate the multi-target anti-colitic effects of CDQD by examining its impact on intestinal homeostasis and its anti-inflammatory properties. METHODS A dextran sulfate sodium (DSS)-induced mouse model of acute colitis was employed. Interferon-gamma (IFN-γ) and KPT-330 were used to induce macrophage PANoptosis. Ultra-high-performance liquid chromatography-high-resolution mass spectrometry (UHPLCHRMS) was utilized to identify the chemical constituents of CDQD. Multi-omics analysis and fecal microbiota transplantation (FMT) were used to explore the therapeutic targets and gut microbiota alterations induced by CDQD. RESULTS CDQD treatment significantly alleviated colitis symptoms in mice, with a dose-dependent therapeutic effect. The decoction mitigated PANoptosis in colon tissues and bone marrow-derived macrophages (BMDMs). 16S rRNA sequencing analysis and metabonomics revealed that CDQD administration significantly altered the gut microbiota composition and metabolite profiles. Notably, CDQD-modulated gut microbiota exhibited anti-colitic effects through FMT. Integrated transcriptomics and network pharmacology analysis revealed that CDQD significantly downregulated the PI3K/Akt signaling pathway in colitis. This finding was further validated using the inhibitors LY294002 and MK2206. CONCLUSIONS CDQD alleviates colitis by suppressing inflammatory macrophage activation and modulating the gut microbiota. Our research provides a novel traditional Chinese medicine strategy for the treatment of UC via enema administration.
Collapse
Affiliation(s)
- Zelong Lin
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120, China; Chinese Medicine Guangdong Laboratory, Guangdong Hengqin, China
| | - Jun Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120, China; Guangdong Provincial Key Laboratory of Chinese Medicine for Prevention and Treatment for Refractory Chronic Diseases, China
| | - Huishan Luo
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120, China
| | - Linwen Huang
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120, China
| | - Zhaoyu Pan
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120, China
| | - Shilong Yang
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120, China
| | - Cailing Zhong
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120, China
| | - Ng Chong Shan
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120, China
| | - Ziwen Ye
- School of Nursing, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Huishi Tan
- Department of Gastroenterology and Hepatology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China
| | - Xiaobo Yang
- Chinese Medicine Guangdong Laboratory, Guangdong Hengqin, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou 510120, China.
| | - Beiping Zhang
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, China.
| | - Chongyang Huang
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120, China.
| | - Haiyan Zhang
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou 510120, China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou 510120, China.
| |
Collapse
|
15
|
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
|
16
|
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
|
17
|
Yang J, Lin D, Liu D, Zhang D, Wang H. A PANoptosis-Based Signature for Survival and Immune Predication in Glioblastoma Multiforme. Ann Clin Transl Neurol 2025. [PMID: 40333895 DOI: 10.1002/acn3.70066] [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: 11/29/2024] [Revised: 03/27/2025] [Accepted: 04/14/2025] [Indexed: 05/09/2025] Open
Abstract
OBJECTIVE PANoptosis is a concept of total cell death characterized by pyroptosis, apoptosis, and necroptosis. We aimed to explore the clinical significance of PANoptosis-related genes (PARGs) in glioblastoma multiforme (GBM). METHODS Expression profiles of GBM were downloaded from the XENA database as a training dataset to construct a differentially expressed PARGs (DE-PARGs)-based risk score (RS) model, and the prognostic prediction role was validated in the CGGA database and GSE108474 using Kaplan-Meier (KM) curve and receiver operating characteristic (ROC) curve. Meanwhile, independent prognostic clinical factors were screened, and their prognosis predictive activity was evaluated by a nomogram model. Furthermore, the relationships between key DE-PARGs and immune cell infiltration, as well as chemotherapy drug sensitivity were analyzed. RESULTS The RS model consisting of five DE-PARGs was constructed, including NOD2, NLRP2, NLRP7, GATA3, and TERT. ROC and KM curves confirmed the good potency of the RS prognostic model both in XENA database and GSE108474. Three clinical prognostic factors, including chemotherapy, pharmaceutical therapy, and RS model, were selected as individual prognostic factors. The nomogram model showed RS contributed most to survival probability, followed by chemotherapy and pharmaceutical therapy. In high- and low-risk groups, B cell memory, NK cell resting, and macrophage M1 had significant differences. As compared with the immune checkpoint therapy non-responder group, the responder involved a higher ratio of patients sub-grouped into the low-risk group. Three drugs between high- and low-risk groups had significant differences, including Cisplatin, Gefitinib, and Vorinostat. INTERPRETATION Our data exhibit the prognostic value of PARGs in GBM and offer new insights for GBM pathogenesis and immune treatment.
Collapse
Affiliation(s)
- Jun Yang
- Department of Neurosurgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Da Lin
- Department of Neurosurgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Dongyuan Liu
- Department of Neurosurgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Dongxu Zhang
- Department of Neurosurgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Hao Wang
- Department of Neurosurgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
18
|
Gao Y, Yang L, Wu H, Yao Q, Wang J, Zheng N. Sodium butyrate attenuates experimental neonatal necrotizing enterocolitis by suppressing TLR4-mediated NLRP3 inflammasome-dependent pyroptosis. Food Funct 2025; 16:3508-3524. [PMID: 40223745 DOI: 10.1039/d4fo03517h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
Necrotizing enterocolitis (NEC) is a fatal intestinal disease in premature infants, and is characterized by intestinal inflammation and disruption of the intestinal barrier. The protective effects of sodium butyrate (NaB) against NEC have been documented, however, the underlying fundamental processes remain unknown. To address this deficit, we used the NEC neonatal rat model to confirm the intestinal protective effect of NaB. We then used network pharmacology and confirmed a role for NaB in the attenuation of NEC and this was associated with the NLRP3 inflammasome and the NF-κB signaling pathway. These results were verified by proteome analysis in vivo, and molecular docking analysis was used to explore the potential underlying mechanisms, revealing a suppressive function of NaB on NEC, which may be caused by its interaction with the TLR4-mediated NF-κB signaling pathway. An in vitro cell model (LPS-stimulated IEC-6 cells) was then established to confirm the docking results. Results using assays involving the NLRP3 (MCC950) and TLR4 (TAK-242) inhibitors suggested that NaB protected intestinal cells from inflammatory injuries during NEC by suppressing the TLR4/MyD88/NF-κB/NLRP3/cleaved caspase-1/GSDMD inflammasome pathway. These findings indicated that NaB can be used as a potential modulatory and therapeutic candidate for the treatment of NEC.
Collapse
Affiliation(s)
- Yanan Gao
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| | - Liting Yang
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Hongya Wu
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| | - Qianqian Yao
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
- Department of Food Science, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Jiaqi Wang
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| | - Nan Zheng
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| |
Collapse
|
19
|
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
|
20
|
Yang JY, Luo CH, Wang KB, Tu XY, Xiao YY, Ou YT, Xie YX, Guan CX, Zhong WJ. Unraveling the mechanisms of NINJ1-mediated plasma membrane rupture in lytic cell death and related diseases. Int J Biol Macromol 2025; 309:143165. [PMID: 40239793 DOI: 10.1016/j.ijbiomac.2025.143165] [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: 01/29/2025] [Revised: 04/03/2025] [Accepted: 04/13/2025] [Indexed: 04/18/2025]
Abstract
Plasma membrane rupture (PMR), the ultimate event during lytic cell death, releases damage-associated molecular patterns (DAMPs) that trigger inflammation and immune responses in the development of various diseases. Recent years have witnessed significant advances in understanding the PMR mediated by ninjurin1 (NINJ1) in different lytic cell death processes. NINJ1 oligomerizes and ruptures the membrane in pyroptosis and other lytic cell death, participating in the pathogenesis of multiple diseases. Although the membrane-permeabilizing function of NINJ1 is well recognized, the role of NINJ1 in different types of lytic cell death and its impact on multiple disease processes have yet to be fully elucidated. This review summarizes the latest advances in the mechanisms of NINJ1-mediated PMR, discusses the membrane-inducing activity of NINJ1 in different lytic cell death, explains the implications of NINJ1 in lytic cell death-related diseases, and lists the inhibitory strategies for NINJ1. We expect to provide new insights into targeting NINJ1 to suppress lytic cell death for therapeutic benefit, which may become a new strategy to control inflammatory cell lysis-related diseases.
Collapse
Affiliation(s)
- Ji-Yan Yang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China; Key Laboratory of the General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China; National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410078, China
| | - Chen-Hua Luo
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Kun-Bo Wang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Xin-Yu Tu
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Yun-Ying Xiao
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Ye-Tong Ou
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Yan-Xin Xie
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China
| | - Cha-Xiang Guan
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China; Key Laboratory of the General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China; National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410078, China
| | - Wen-Jing Zhong
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410078, China; Key Laboratory of the General University of Hunan Province, Basic and Clinic Research in Major Respiratory Disease, Changsha, Hunan 410078, China; National Experimental Teaching Demonstration Center for Medical Function, Changsha, Hunan 410078, China.
| |
Collapse
|
21
|
Wu J, Guo Z, Wang L, Shen Y, Li X, Zhang Z, Han X, Zhang J, Cai K, Tang C. Porphyromonas gingivalis induces Zbp1-mediated macrophages PANoptosis in periodonitis pathophysiology. Exp Mol Med 2025:10.1038/s12276-025-01443-y. [PMID: 40307566 DOI: 10.1038/s12276-025-01443-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 01/18/2025] [Accepted: 02/09/2025] [Indexed: 05/02/2025] Open
Abstract
Periodontitis is an oral immunoinflammatory disease, and macrophages play a crucial role in its pathophysiology. However, macrophage death during antibacterial activities will exacerbate inflammation and tissue damage. Porphyromonas gingivalis is a major constituent of subgingival biofilm plaques in periodontitis, but the effects and precise molecular mechanisms by which it triggers macrophage death remain unknown. Here we found that P. gingivalis infection notably activated multiple death pathways in bone-marrow-derived macrophages, including pyroptosis, apoptosis and necrosis. Furthermore, using RNA sequencing, we identified that P. gingivalis infection markedly increased the expression of Z-DNA binding protein 1 (Zbp1) in bone-marrow-derived macrophages. Initially identified as an interferon-induced tumor-associated protein, Zbp1 serves as an upstream sensor that regulates cell death by activating PANoptosis. Mechanistically, P. gingivalis induced a mitochondrial stress response, prompting the release of mitochondrial DNA. This mitochondrial DNA then interacted with Zbp1, consequently augmenting its downstream PANoptosis signals. In addition, P. gingivalis stimulated macrophage Zbp1 expression through the Tlr2/4-JNK-Stat3/5 pathway, exacerbating macrophage death. Importantly, blocking the biosynthesis of endogenous Zbp1 by pharmacological delivery with microneedles improved the survival of P. gingivalis-infected macrophages and inhibited periodontal tissue destruction. These findings highlight Zbp1 as a potential therapeutic target for P. gingivalis-induced periodontitis.
Collapse
Affiliation(s)
- Jin Wu
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Oral Implantology Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Zixiang Guo
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Oral Implantology Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Long Wang
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Oral Implantology Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Yue Shen
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Oral Implantology Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Xiang Li
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Oral and Maxillofacial Surgery Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Zhewei Zhang
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Oral Implantology Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Xiao Han
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing, China
- Department of Oral Implantology Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Jianlan Zhang
- Department of Oral Implantology Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
| | - Kunzhan Cai
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing, China.
- Department of Oral Implantology Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China.
| | - Chunbo Tang
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Nanjing, China.
- Department of Oral Implantology Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
22
|
Liu Z, Shi J, Tu K, Ma H, Chen J, Xiang X, Zou P, Liao C, Ding R, Huang Z, Yao X, Chen J, Wang L, Zhang Z. GPx3 Promotes Functional Recovery after Spinal Cord Injury by Inhibiting Microglial Pyroptosis Through IRAK4/ROS/NLRP3 Axis. Antioxid Redox Signal 2025; 42:711-729. [PMID: 39895340 DOI: 10.1089/ars.2024.0618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Aim: Spinal cord injury (SCI) is a catastrophic injury characterized by oxidative stress. Glutathione peroxidase 3 (GPx3) is an antioxidant enzyme that protects against immune responses in various diseases. However, the effects of GPx3 in SCI remains unclear. This study aimed to investigate the role of GPx3 in SCI and its underlying mechanisms. Results: We injected adeno-associated viruses to overexpress GPx3 in mice. Primary microglia and BV2 cells were used as in vitro models. We knocked down or overexpressed GPx3 in BV2 cells. Additionally, BV2 cells transfected with siIRAK4 were used to perform rescue experiments. A series of histological and molecular biological analyses were used to explore the role of GPx3 in SCI. Overexpression of GPx3 inhibited oxidative stress in mice, improving functional recovery after SCI. Similarly, LPS+ATP stimulation decreased GPx3 expression in microglia. Silencing of GPx3 elevated the generation of reactive oxygen species, increased the expression of IRAK4 and pro-inflammatory factors, and promoted pyroptosis in microglia. However, overexpression of GPx3 reversed these results. Moreover, silencing of IRAK4 alleviated these phenomena, which were upregulated by GPx3 deficiency. Innovation and Conclusion: Our results demonstrated that GPx3 plays a critical role in SCI by inhibiting microglial pyroptosis via the IRAK4/ROS/NLRP3 signaling pathway. Antioxid. Redox Signal. 42, 711-729.
Collapse
Affiliation(s)
- Zhongyuan Liu
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiawei Shi
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Kewu Tu
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hao Ma
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiayu Chen
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xin Xiang
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Peiqian Zou
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Congrui Liao
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ruoting Ding
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zucheng Huang
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xinqiang Yao
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jianting Chen
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Liang Wang
- Department of Orthopaedics, Academy of Orthopedics, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Zhongmin Zhang
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
23
|
Wang Y, Yuan M, Qin J, Chen X, Lei Z, Kong Q, Wang Q, Song X, Wu X. TIPE2 Alleviates Sepsis-induced Lung Injury By Inhibiting PANoptosis in Murine Alveolar Macrophages. Inflammation 2025:10.1007/s10753-025-02288-3. [PMID: 40299241 DOI: 10.1007/s10753-025-02288-3] [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: 12/01/2024] [Revised: 01/20/2025] [Accepted: 03/05/2025] [Indexed: 04/30/2025]
Abstract
Sepsis-induced acute lung injury (ALI) is a life-threatening condition with high mortality rates, and its underlying mechanisms remain poorly understood. This study investigates the role of TNF-α-induced protein 8-like 2 (TIPE2) in modulating PANoptosis, an integrated form of programmed cell death that includes apoptosis, necroptosis, and pyroptosis, in the context of sepsis-induced lung injury. We utilized a cecal ligation and puncture (CLP) mouse model to examine the effects of TIPE2 knockout and overexpression on lung injury, inflammation, and cell death pathways. Our findings demonstrate that TIPE2 knockout exacerbates lung injury by promoting the abnormal activation of PANoptosis-related proteins, leading to increased inflammation and tissue damage. In contrast, overexpression of TIPE2 in macrophages significantly reduces these effects by inhibiting the ZBP1-dependent PANoptosis pathway via TRIF signaling. These results highlight the crucial role of TIPE2 in maintaining the balance between cell survival and death during sepsis and suggest that targeting TIPE2 could be a novel therapeutic strategy for treating sepsis-related lung injury.
Collapse
Affiliation(s)
- Yuxuan Wang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Min Yuan
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jingxue Qin
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xue Chen
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Zihan Lei
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Qian Kong
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Qian Wang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xuemin Song
- Research Centre of Anesthesiology and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China.
| | - Xiaojing Wu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
24
|
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
|
25
|
Wang D, Yuan Y, Tong X, Wang L, Sun J, Zhang S, Liu S, Gan H, Fan H. Identification and Experimental Validation of PANoptosis-Related Genes in Idiopathic Pulmonary Fibrosis by Bioinformatics Analysis. J Inflamm Res 2025; 18:5499-5517. [PMID: 40291456 PMCID: PMC12034268 DOI: 10.2147/jir.s505229] [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: 11/25/2024] [Accepted: 04/01/2025] [Indexed: 04/30/2025] Open
Abstract
Aim To identify the molecular signature of differentially expressed genes (DEGs) associated with PANoptosis in idiopathic pulmonary fibrosis (IPF) and to interpret their immune landscape and cellular distribution characteristics. Methods and Results We acquired two IPF datasets from the Gene Expression Omnibus (GEO) database to identify PANoptosis-related DGEs (PAN-DEGs), initially identifying thirty PAN-DEGs. Utilizing machine learning algorithms, we established a five-gene PANoptosis-related signature comprising IGF1, GPX3, GADD45β, SMAD7, and TIMP3, each demonstrating robust diagnostic performance. The expression of these hub genes was subsequently validated using a third GEO dataset and a bleomycin-induced pulmonary fibrosis model. Immune infiltration analysis revealed a close association of these genes with various immune cells, and single-cell RNA sequencing indicated significant expression changes in diverse pulmonary cell types, particularly endothelial cells and fibroblasts. Conclusion We identified and validated a PANoptosis-related gene signature in IPF, providing insights into their immune infiltration and potential cellular distribution. Further research is necessary to elucidate the biological functions and mechanisms of these genes in the pathogenesis of IPF.
Collapse
Affiliation(s)
- Dongguang Wang
- Department of Respiratory and Critical Care Medicine, and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
- State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Yifan Yuan
- Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
- Centre for Inflammatory Bowel Disease, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
- Lab of Inflammatory Bowel Disease, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Xiang Tong
- Department of Respiratory and Critical Care Medicine, and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
- State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Lian Wang
- Department of Respiratory and Critical Care Medicine, and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
- State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Jibo Sun
- Department of Respiratory and Critical Care Medicine, and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
- State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Shijie Zhang
- Department of Respiratory and Critical Care Medicine, and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
- State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Sitong Liu
- Department of Respiratory and Critical Care Medicine, and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
- State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Huatian Gan
- Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
- Centre for Inflammatory Bowel Disease, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
- Lab of Inflammatory Bowel Disease, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Hong Fan
- Department of Respiratory and Critical Care Medicine, and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
- State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| |
Collapse
|
26
|
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
|
27
|
Zhou J, Wu Z, Zhao P. Luteolin and its antidepressant properties: From mechanism of action to potential therapeutic application. J Pharm Anal 2025; 15:101097. [PMID: 40276566 PMCID: PMC12018562 DOI: 10.1016/j.jpha.2024.101097] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 07/18/2024] [Accepted: 09/03/2024] [Indexed: 04/26/2025] Open
Abstract
Luteolin is a natural flavonoid compound exists in various fruits and vegetables. Recent studies have indicated that luteolin has variety pharmacological effects, including a wide range of antidepressant properties. Here, we systematically review the preclinical studies and limited clinical evidence on the antidepressant and neuroprotective effects of luteolin to fully explore its antidepressant power. Network pharmacology and molecular docking analyses contribute to a better understanding of the preclinical models of depression and antidepressant properties of luteolin. Seventeen preclinical studies were included that combined network pharmacology and molecular docking analyses to clarify the antidepressant mechanism of luteolin and its antidepressant targets. The antidepressant effects of luteolin may involve promoting intracellular noradrenaline (NE) uptake; inhibiting 5-hydroxytryptamine (5-HT) reuptake; upregulating the expression of synaptophysin, postsynaptic density protein 95, brain-derived neurotrophic factor, B cell lymphoma protein-2, superoxide dismutase, and glutathione S-transferase; and decreasing the expression of malondialdehyde, caspase-3, and amyloid-beta peptides. The antidepressant effects of luteolin are mediated by various mechanisms, including anti-oxidative stress, anti-apoptosis, anti-inflammation, anti-endoplasmic reticulum stress, dopamine transport, synaptic protection, hypothalamic-pituitary-adrenal axis regulation, and 5-HT metabolism. Additionally, we identified insulin-like growth factor 1 receptor (IGF1R), AKT serine/threonine kinase 1 (AKT1), prostaglandin-endoperoxide synthase 2 (PTGS2), estrogen receptor alpha (ESR1), and epidermal growth factor receptor (EGFR) as potential targets, luteolin has an ideal affinity for these targets, suggesting that it may play a positive role in depression through multiple targets, mechanisms, and pathways. However, the clinical efficacy of luteolin and its potential direct targets must be confirmed in further multicenter clinical case-control and molecular targeting studies.
Collapse
Affiliation(s)
- Jiayu Zhou
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Ziyi Wu
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Ping Zhao
- Department of Anesthesiology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| |
Collapse
|
28
|
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
|
29
|
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
|
30
|
Mealy JE, Torres WM, Freeburg LA, Barlow SC, Whalen AA, Maduka CV, Shazly TM, Burdick JA, Spinale FG. Shear-Thinning Hydrogel for Delayed Delivery of a Small Molecule Metalloproteinase Inhibitor Attenuates Myocardial Infarction Remodeling. JACC Basic Transl Sci 2025; 10:458-472. [PMID: 40306854 DOI: 10.1016/j.jacbts.2024.11.009] [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: 03/12/2024] [Revised: 10/30/2024] [Accepted: 11/18/2024] [Indexed: 05/02/2025]
Abstract
Strategic delivery of hydrogels to the newly formed myocardial infarction (MI) is an area of active investigation and offers high target specificity for releasing a small molecule therapeutic payload. This study examined the effects of delayed post-MI delivery (pigs, 3 days post-MI) of a shear-thinning hydrogel which encapsulated and released a small molecule matrix metalloproteinase inhibitor. The results demonstrated the feasibility and efficacy of targeted delivery of a shear-thinning injectable hydrogel containing a small molecule matrix metalloproteinase inhibitor to attenuate post-MI remodeling.
Collapse
Affiliation(s)
- Joshua E Mealy
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - William M Torres
- College of Engineering and Computing, University of South Carolina, Columbia, South Carolina, USA; Cardiovascular Translational Research Center, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Lisa A Freeburg
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Shayne C Barlow
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Alison A Whalen
- Cardiovascular Translational Research Center, University of South Carolina School of Medicine, Columbia, South Carolina, USA
| | - Chima V Maduka
- BioFrontiers Institute and Department of Chemical and Biological Engineering, University of Colorado-Boulder, Boulder, Colorado, USA
| | - Tarek M Shazly
- College of Engineering and Computing, University of South Carolina, Columbia, South Carolina, USA
| | - Jason A Burdick
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, USA; BioFrontiers Institute and Department of Chemical and Biological Engineering, University of Colorado-Boulder, Boulder, Colorado, USA
| | - Francis G Spinale
- College of Engineering and Computing, University of South Carolina, Columbia, South Carolina, USA; Cardiovascular Translational Research Center, University of South Carolina School of Medicine, Columbia, South Carolina, USA; Columbia VA Health Care Center, Columbia, South Carolina, USA.
| |
Collapse
|
31
|
Xia X, Kong C, Zhao X, Zhao K, Shi N, Jiang J, Li P. The complexities of cell death mechanisms: a new perspective in systemic sclerosis therapy. Apoptosis 2025; 30:636-651. [PMID: 39924583 DOI: 10.1007/s10495-025-02082-4] [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] [Accepted: 01/21/2025] [Indexed: 02/11/2025]
Abstract
Systemic sclerosis, also termed scleroderma, is a severe and debilitating autoimmune disease characterized by fibrosis, an aberrant immune response, and vascular dysfunction. Cell death is essential to the body's continued normal development as it removes old or damaged cells. This process is governed by several mechanisms, including programmed cell death through apoptosis, necrosis, and pyroptosis, as well as metabolic processes, such as ferroptosis and cuproptosis. This review describes the signaling pathways associated with each form of cell death, examining the linkages between these pathways, and discussing how the dysregulation of cell death processes is involved in the development of autoimmune disorders such as systemic sclerosis. Existing and promising therapeutic strategies aimed at restoring the balance of cell death in systemic sclerosis and other autoimmune disorders are also emphasized.
Collapse
Affiliation(s)
- Xue Xia
- Department of Rheumatology and Immunology, China-Japan Union Hospital, Jilin University, Changchun, 130033, China
| | - Chenfei Kong
- Scientific Research Center, China-Japan Union Hospital, Jilin University, Changchun, 130033, China
| | - Xiaoming Zhao
- Scientific Research Center, China-Japan Union Hospital, Jilin University, Changchun, 130033, China
| | - Kelin Zhao
- Department of Rheumatology and Immunology, China-Japan Union Hospital, Jilin University, Changchun, 130033, China
| | - Naixu Shi
- Department of Stomatology, China-Japan Union Hospital of Jilin University, Changchun, 130033, China
| | - Jinlan Jiang
- Scientific Research Center, China-Japan Union Hospital, Jilin University, Changchun, 130033, China.
| | - Ping Li
- Department of Rheumatology and Immunology, China-Japan Union Hospital, Jilin University, Changchun, 130033, China.
| |
Collapse
|
32
|
Wang Z, Yang Y, Yao FT, Zhang F, Lin KY, Diao HT, Zhao QY, Kong X, Si W, Xie YT, Song JL, Zeng LH, Wang CL, Xiong YT, Zou KK, Wang XM, Zhang XY, Wu H, Jiang WT, Bian Y, Yang BF. KLX ameliorates liver cancer progression by mediating ZBP1 transcription and ubiquitination and increasing ZBP1-induced PANoptosis. Acta Pharmacol Sin 2025:10.1038/s41401-025-01528-4. [PMID: 40148674 DOI: 10.1038/s41401-025-01528-4] [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: 09/24/2024] [Accepted: 02/26/2025] [Indexed: 03/29/2025]
Abstract
Liver cancer is a highly aggressive malignancy with poor survival rates. Current treatments, including liver transplantation, immunotherapy, and gene therapy, are often limited by late-stage diagnosis and significant side effects, highlighting the urgent need for novel therapeutic agents. In this study, we evaluated the therapeutic potential of Kanglexin (KLX), a novel anthraquinone derivative, in the treatment of liver cancer. In vitro, KLX inhibited the proliferation and migration of HepG2 and Hep3B cells in a dose-dependent manner. Mechanistically, KLX upregulated Z-DNA binding protein 1 (ZBP1) expression, inducing PANoptosis by directly binding to ZBP1, altering its conformation, and reducing its affinity for the E3 ubiquitin ligase ring finger protein 180 (RNF180). This interaction decreased ZBP1 ubiquitination, thereby increasing its stability. Additionally, KLX upregulated the expression of the transcription factor homeobox D10 (HOXD10), which further increased ZBP1 expression. Elevated ZBP1 levels significantly suppressed liver cancer cell proliferation and migration, whereas the inhibitory effects of KLX were reversed upon ZBP1 knockdown. In a xenograft model, KLX significantly inhibited tumor growth with a lower toxicity than oxaliplatin (OXA). In conclusion, KLX promoted PANoptosis in liver cancer cells by upregulating ZBP1 and preventing its degradation, thereby inhibiting liver cancer progression and migration. These findings suggest that KLX is a promising therapeutic agent for liver cancer.
Collapse
Affiliation(s)
- Zhuo Wang
- College of Traditional Chinese Medicine and Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yang Yang
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Fang-Ting Yao
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Feng Zhang
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Ke-Ying Lin
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Hong-Tao Diao
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Qiao-Yue Zhao
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Xue Kong
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Wei Si
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Ya-Ting Xie
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Jing-Lun Song
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Ling-Hua Zeng
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Chun-Lei Wang
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Yu-Ting Xiong
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Kun-Kun Zou
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Xiao-Man Wang
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Xin-Yue Zhang
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Han Wu
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Wei-Tao Jiang
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China
| | - Yu Bian
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China.
| | - Bao-Feng Yang
- College of Traditional Chinese Medicine and Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
- Department of Pharmacology (National Key Laboratory of Frigid Zone Cardiovascular Diseases, the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, 150081, China.
| |
Collapse
|
33
|
Yan T, Nan J, Jiang R, Chen F, Li J. Nd:YAG1064nm laser functions against Sporothrix globosa by inducing PANoptosis via the regulation of ZBP1-induced PANoptosome activation. Front Microbiol 2025; 16:1555338. [PMID: 40207151 PMCID: PMC11979269 DOI: 10.3389/fmicb.2025.1555338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 03/14/2025] [Indexed: 04/11/2025] Open
Abstract
Background Due to the emergence of drug resistance in recent years, there is a need for new non-pharmacological treatment methods for sporotrichosis. Our previous study demonstrated that the Nd:YAG1064nm laser exhibited remarkable antifungal activity against Sporothrix globosa, but its exact mechanism remains unclear. This study aimed to detect PANoptosis regulatory protein ZBP1 expression in the skin lesions of patients with sporotrichosis, reveal the exact mechanism of Nd:YAG1064nm laser against sporotrichosis, and provide novel targets and methods for the diagnosis, assessment, and treatment of sporotrichosis. Methodology/principal findings The ZBP1 level of 60 patients with sporotrichosis (≤3 months; n = 30 and >3 months; n = 30) and 30 HC were retrospectively reviewed using immunohistochemistry. The morphological changes, Hoechst/PI apoptosis and necroptosis preliminary exploration analysis, DNA fragmentation, calcium determination, and metacaspase activation were investigated in vitro. For the in vivo studies, mice were infected with S. globosa and then treated with a laser, and their footpad skin lesions and changes in the histology of tissue samples were compared. Changes in the levels of ZBP1, PANoptosome [RIPK1, RIPK3, Fas-associated death domain protein (FADD), CASP8], pyroptosis (CASP1, GSDMD), apoptosis (CASP3), and necroptosis (MLKL) related proteins were assessed using immunohistochemistry, whereas the levels of interleukin 17 (IL-17) and interferon gamma (IFN-γ) in peripheral blood were tested by enzyme-linked immunosorbent assay. ZBP1 expression was significantly increased in S. globosa-infected patients. Laser treatment effectively inhibited the growth of S. globosa in vitro, destroying its morphological structure, and maybe inducing apoptosis and necroptosis. Moreover, DNA fragmentation, calcium release into the cytoplasm, and metacaspase activation were observed. In addition, laser treatment demonstrated a clear therapeutic effect in animal models of sporotrichosis, which can lead to PANoptosis-related apoptosis, pyroptosis, and necroptosis. Immune response-related macrophages perceive nucleic acid level changes through ZBP1 to recognize S. globosa and induce PANoptosis by activating the PANoptosome (RIPK1/RIPK3/FADD/CASP8) complex with a Th1/Th17 cell response to combat sporotrichosis. Conclusion Nd:YAG1064nm laser mediated PANoptosis resistance to sporotrichosis via ZBP1-PANoptosome-PANoptosis pathway.
Collapse
Affiliation(s)
- Tianyi Yan
- Department of Dermatology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jinyan Nan
- Department of Dermatology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Rihua Jiang
- Department of Dermatology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Feng Chen
- Department of Dermatology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jinran Li
- Department of Dermatology, Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
34
|
Wang D, Dai Z, Jiang L, Liu K. Circulating extracellular vesicles regulate ELAVL1 by delivering miR-133a-3p which affecting NLRP3 mRNA stability inhibiting PANoptosome formation. Biol Direct 2025; 20:36. [PMID: 40140903 PMCID: PMC11948929 DOI: 10.1186/s13062-025-00605-2] [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: 08/26/2024] [Accepted: 01/13/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND In the quest to elucidate novel therapeutic strategies for myocardial injury, recent investigations have underscored the pivotal roles played by circulating extracellular vesicles (EVs) in intercellular communication. METHOD EVs were extracted from individuals who had experienced AMI-EVs and those who were N-EVs. To assess the impact of circulating EVs on cardiomyocyte and endothelial cell proliferation, apoptosis, migration, and tube formation, a range of in vitro assays such as CCK8, EdU assays, flow cytometry, wound healing assays and angiogenesis assays were conducted. Differentially expressed miRNAs in EVs were validated using microarray analysis and real-time PCR. Through bioinformatics analysis, ELAVL1 was identified as a potential downstream target of miR-133a-3p. This finding was further confirmed by conducting dual-luciferase reporter assay and RNA co-immunoprecipitation experiments. To investigate the regulatory effects of circulating EVs from various sources on myocardial injury and PANoptosis, an animal model of ischemia-reperfusion-induced myocardial injury was established. RESULT Our findings revealed that circulating EVs effectively deliver miR-133a-3p to target cells, where it binds to ELAVL1, leading to a decrease in NLRP3 mRNA stability. This reduction in NLRP3 mRNA stability subsequently inhibits the assembly of the PANoptosome, a multi-protein complex implicated in PANoptosis. As a result, we observed a significant mitigation of PANoptosis in our myocardial injury models, demonstrating the protective role of miR-133a-3p against excessive cell death. CONCLUSION The present study underscores the regulatory role of circulating EV-delivered miR-133a-3p in modulating PANoptosis through ELAVL1-mediated NLRP3 mRNA stabilization. This mechanism represents a potential therapeutic target for attenuating myocardial injury by suppressing PANoptosis.
Collapse
Affiliation(s)
- Deliang Wang
- Department of Cardiac Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China
| | - Zheng Dai
- Department of Emergency Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China
| | - Lu Jiang
- Department of Cardiac Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China.
| | - Ke Liu
- Department of Cardiac Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, 610072, China.
| |
Collapse
|
35
|
Xie J, Fu Q, Qin L, Lin L, Wu Q, Zeng K, Wu J, Cao Z, Ou Y. Ochratoxin A induces lung cell PANoptosis through activation of the AIM 2 inflammasome. Int Immunopharmacol 2025; 150:114184. [PMID: 39938165 DOI: 10.1016/j.intimp.2025.114184] [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: 11/10/2024] [Revised: 01/22/2025] [Accepted: 01/26/2025] [Indexed: 02/14/2025]
Abstract
Ochratoxin A (OTA), a mycotoxin from filamentous fungi, significantly threatens human and animal health through food contamination. OTA is prevalent in food products, posing a significant health risk. Here, we observed that OTA induces senescence in lung cells. This study further assessed the toxicological effects of OTA on lung cells and clarified its molecular mechanism. We utilized in vitro cell models (TC-1 and MLE-12) to evaluate the impact of OTA on lung cells using Western-blot, indirect immunofluorescence and ELISA. The results revealed that OTA leads to inflammatory cell death in lung cells. Further investigations demonstrated that OTA elevates the expression levels of PANoptosis markers, including ZBP1, Caspase1/GSDMD (pyroptosis), Caspase3/7 (apoptosis) and RIP3/pMLKL (necroptosis). We further explored the mechanism through which OTA induces PANoptosis in lung cells. Experimental results indicated that OTA increased mitochondrial ROS levels, subsequently leading to a decrease in mitochondrial membrane potential, which activates AIM2. Consequently, AIM2 participates in the formation of ZBP1-induced PAN-optosome, ultimately resulting in PANoptosis of lung cells. In vivo studies further revealed that OTA induces lung damage. This new discovery establishes a basis for future studies on the toxicological effects of OTA on lung tissue.
Collapse
Affiliation(s)
- Jianlong Xie
- Department of Thoracic Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, China
| | - Qiujuan Fu
- Pathological Diagnosis and Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, China
| | - Lingling Qin
- Pathological Diagnosis and Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, China; First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong 524000, China
| | - Liyao Lin
- Department of Thoracic Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, China
| | - Qin Wu
- Department of Thoracic Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, China
| | - Kaiqi Zeng
- Pathological Diagnosis and Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, China; First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong 524000, China
| | - Jiahuan Wu
- Pathological Diagnosis and Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, China; First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong 524000, China
| | - Ziyi Cao
- Pathological Diagnosis and Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, China; First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong 524000, China
| | - Yongfang Ou
- Pathological Diagnosis and Research Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524000, China.
| |
Collapse
|
36
|
Xiao J, Wang L, Zhang B, Hou A. Cell death in acute lung injury: caspase-regulated apoptosis, pyroptosis, necroptosis, and PANoptosis. Front Pharmacol 2025; 16:1559659. [PMID: 40191423 PMCID: PMC11968751 DOI: 10.3389/fphar.2025.1559659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 03/03/2025] [Indexed: 04/09/2025] Open
Abstract
There has been abundant research on the variety of programmed cell death pathways. Apoptosis, pyroptosis, and necroptosis under the action of the caspase family are essential for the innate immune response. Caspases are classified into inflammatory caspase-1/4/5/11, apoptotic caspase-3/6/7, and caspase-2/8/9/10. Although necroptosis is not caspase-dependent to transmit cell death signals, it can cross-link with pyroptosis and apoptosis signals under the regulation of caspase-8. An increasing number of studies have reiterated the involvement of the caspase family in acute lung injuries caused by bacterial and viral infections, blood transfusion, and ventilation, which is influenced by noxious stimuli that activate or inhibit caspase engagement pathways, leading to subsequent lung injury. This article reviews the role of caspases implicated in diverse programmed cell death mechanisms in acute lung injury and the status of research on relevant inhibitors against essential target proteins of the described cell death mechanisms. The findings of this review may help in delineating novel therapeutic targets for acute lung injury.
Collapse
Affiliation(s)
| | | | | | - Ana Hou
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
37
|
Hu H, Chen J, Shao Y, Tang Y, Dun Y, Memet O, Bao X, Shen J. Establishment of rat model for aspiration pneumonia and potential mechanisms. Animal Model Exp Med 2025. [PMID: 40110746 DOI: 10.1002/ame2.12566] [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: 07/31/2024] [Accepted: 01/05/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Aspiration pneumonia is a severe health concern, particularly for ICU patients with impaired airway defenses. Current animal models fail to fully replicate the condition, focusing solely on chemical lung injury from gastric acid while neglecting pathogen-induced inflammation. This gap hinders research on pathogenesis and treatment, creating an urgent need for a clinically relevant model. This study aimed to develop an improved rat model of aspiration pneumonia by combining hydrochloric acid (HCl) and lipopolysaccharide (LPS) administration. METHODS Specific pathogen-free Sprague Dawley rats underwent intratracheal instillation of HCl and LPS. Techniques included rat weight measurement, tracheal intubation, pulmonary function monitoring, lung tissue sampling with HE staining and scoring, bronchoalveolar lavage fluid (BALF) sampling, protein and inflammatory cytokine analysis via BCA and ELISA, BALF pH determination, Evans Blue dye assessment, blood gas analysis, FITC-dextran leakage, Western blotting, electron microscopy, survival analysis, and transcriptome sequencing with bioinformatics. Statistical analysis was performed using GraphPad Prism. RESULTS The optimal model involved instillation of 1.5 μL/g.wt HCl (pH = 1) followed by 20 μg/g.wt LPS after 1 h. This model reproduced acute lung injury, including tissue damage, pulmonary microvascular dysfunction, inflammatory responses, hypoxemia, and impaired pulmonary ventilation, with recovery observed at 72 h. PANoptosis was confirmed, characterized by increased markers. Concentration-dependent effects of HCl and LPS on lung damage were identified, alongside cytokine elevation and microvascular dysfunction. CONCLUSIONS This optimized model closely mimics clinical aspiration pneumonia, providing a valuable tool for studying pathophysiology and therapeutic strategies.
Collapse
Affiliation(s)
- Hanbing Hu
- Center of Emergency and Critical Medicine, Jinshan Hospital of Fudan University, Shanghai, China
- Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai, China
- Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai, China
| | - Junfeng Chen
- Center of Emergency and Critical Medicine, Jinshan Hospital of Fudan University, Shanghai, China
- Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai, China
- Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai, China
| | - Yiru Shao
- Center of Emergency and Critical Medicine, Jinshan Hospital of Fudan University, Shanghai, China
- Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai, China
- Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai, China
| | - Yuedong Tang
- Center of Emergency and Critical Medicine, Jinshan Hospital of Fudan University, Shanghai, China
- Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai, China
- Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai, China
| | - Yu Dun
- Center of Emergency and Critical Medicine, Jinshan Hospital of Fudan University, Shanghai, China
- Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai, China
- Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai, China
| | - Obulkasim Memet
- Center of Emergency and Critical Medicine, Jinshan Hospital of Fudan University, Shanghai, China
- Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai, China
- Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai, China
| | - Xuanrong Bao
- Center of Emergency and Critical Medicine, Jinshan Hospital of Fudan University, Shanghai, China
- Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai, China
- Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai, China
| | - Jie Shen
- Center of Emergency and Critical Medicine, Jinshan Hospital of Fudan University, Shanghai, China
- Research Center for Chemical Injury, Emergency and Critical Medicine of Fudan University, Shanghai, China
- Key Laboratory of Chemical Injury, Emergency and Critical Medicine of Shanghai Municipal Health Commission, Shanghai, China
| |
Collapse
|
38
|
Li J, Zhu C, Meng Y, Zhang L, Liu C, Qin Y, Chen M. Zika virus inhibits cell death by inhibiting the expression of NLRP3 and A20. J Virol 2025; 99:e0198024. [PMID: 39976465 PMCID: PMC11915814 DOI: 10.1128/jvi.01980-24] [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: 11/07/2024] [Accepted: 01/20/2025] [Indexed: 03/19/2025] Open
Abstract
Zika virus (ZIKV) is associated with microcephaly in neonates and neurological disorders in adults. Chronic ZIKV infection has been identified in the testes, indicating that the virus can lead to prolonged illness, yet its pathogenesis remains poorly understood. Here, we found that ZIKV infection does not induce significant cell death in mouse macrophages despite the critical role that cell death plays in the antiviral immune response. Furthermore, we discovered that ZIKV infection impairs the activation of the NLPR3-dependent inflammasome and inhibits apoptosis. Consequently, we investigated the regulatory mechanism of the NLRP3 inflammasome and apoptosis in the context of ZIKV infection. Our results revealed significant reductions in the protein expression levels of NLRP3 and A20, attributable to post-transcriptional or translational effects during ZIKV infection. These findings suggest that ZIKV infection may disrupt cell death pathways, leading to its pathogenicity.IMPORTANCEZika virus (ZIKV), first isolated from a nonhuman primate in Africa in 1947, was relatively understudied until 2016. By then, ZIKV had already been reported in more than 20 countries and territories. The infection poses a significant risk, as it is associated with microcephaly in infants and neurological disorders in adults; however, the underlying mechanisms responsible for these severe outcomes remain unclear. In this study, we demonstrate that ZIKV infection significantly reduces the expression of NLRP3 and A20 proteins through post-transcriptional or translational processes, which leads to inhibited cell death. These findings are critical because cell death plays a vital role in the host's antiviral immune response. Our findings highlight how ZIKV infection compromises essential cell death pathways, raising serious concerns about its pathogenesis. A comprehensive understanding of this disruption is vital for developing targeted interventions to mitigate the virus' impact on public health.
Collapse
Affiliation(s)
- Jian Li
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Changyang Zhu
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Yang Meng
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Linliang Zhang
- College of Life Sciences, Hubei University, Wuhan, China
| | - Cong Liu
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Yali Qin
- College of Life Sciences, Hubei University, Wuhan, China
| | - Mingzhou Chen
- State Key Laboratory of Virology and Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
- College of Life Sciences, Hubei University, Wuhan, China
- Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- Hubei Jiangxia Laboratory, Wuhan, China
| |
Collapse
|
39
|
Zeng Z, Liu X, Xiang F, He X, Li J, Liu H, Xie L. MicroRNA-21 plays a role in exacerbating chronic obstructive pulmonary disease by regulating necroptosis and apoptosis in bronchial epithelial cells. Tob Induc Dis 2025; 23:TID-23-32. [PMID: 40104397 PMCID: PMC11915094 DOI: 10.18332/tid/202182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 10/31/2024] [Accepted: 02/22/2025] [Indexed: 03/20/2025] Open
Abstract
INTRODUCTION Bronchial epithelial cell damage is an important determinant of the severity of chronic obstructive pulmonary (COPD). However, the exact molecular mechanisms underlying this cell death in COPD development are not well understood. This study investigates the involvement of microRNA-21 (miR-21/miRNA-21) in COPD and its underlying molecular mechanism. METHODS A mouse model of COPD was created by exposing the mice to cigarette smoke (CS) and injecting them with cigarette smoke extract (CSE). Both wild-type mice and miR-21 knockout (miR-21-/-) mice were used to investigate the role of microRNA-21 (miR-21) in exacerbating COPD. Various assays and analyses were performed, including HE staining, tunel staining, enzyme-linked immunosorbent assay (ELISA), flow cytometry, quantitative real-time polymerase chain reaction (RT-qPCR), and western blotting (WB) to measure outcomes such as the pathological morphological changes, necroptosis, apoptosis, and levels of inflammatory factors. RESULTS Our results revealed an upregulation of miR-21 in the lung tissue of COPD model mice. Additionally, knockout of miR-21 resulted in decreased levels of bronchial epithelial cell necroptosis and apoptosis, as evidenced by the downregulation of tumor necrosis factor receptor 1 (TNFR1), phosphoryl-mixed lineage kinase domain-like protein (p-MLKL) and caspase-3. This downregulation of necroptosis and apoptosis ultimately led to a reduction of inflammatory factors and damage-associated molecular patterns (DAMPs), such as tumor necrosis factor-α (TNF-α), interleukin-1β (IL- 1β), and interleukin-6 (IL-6) and high mobility group protein B1(HMGB1) in the lungs, thereby ameliorating COPD. CONCLUSIONS Our findings suggest that miR-21 contributes to the worsening of chronic obstructive pulmonary disease by modulating necroptosis and apoptosis in bronchial epithelial cells, providing a new theoretical basis for the pathogenesis of chronic obstructive pulmonary disease.
Collapse
Affiliation(s)
- Zhengpeng Zeng
- Health Management Medicine Center, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xuelian Liu
- Health Management Medicine Center, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Fei Xiang
- Respiratory and Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Xue He
- Health Management Medicine Center, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Jiahui Li
- Changsha Kexin Cancer Hospital, Changsha, China
| | - Hanying Liu
- Health Management Medicine Center, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Lihua Xie
- Respiratory and Critical Care Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
40
|
Zhuang S, Li F, Wang L, Lai Z, Li D, Wu H, Wu J, Qu J, Zhang X, Zhang M, Chen R, Yuan X. Neutrophil extracellular trap-derived double-stranded RNA aggravates PANoptosis in renal ischemia reperfusion injury. Cell Commun Signal 2025; 23:140. [PMID: 40098148 PMCID: PMC11912734 DOI: 10.1186/s12964-025-02145-8] [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: 09/18/2024] [Accepted: 03/08/2025] [Indexed: 03/19/2025] Open
Abstract
A dysregulated inflammatory response and inflammation-associated cell death are central features of renal ischemia-reperfusion injury (IRI). PANoptosis, is a recently recognized form of inflammatory programmed cell death characterized by key features of pyroptosis, apoptosis and necroptosis; however, the specific involvement of PANoptosis in renal IRI remains unknown. By using neutrophil extracellular trap (NETs)-depleted Pad4-/- mice, we found that NETs are essential for exacerbating tissue injury in renal IRI. Single-cell RNA sequencing (scRNA-seq) revealed that IRI promoted PANoptosis signalling in proximal tubular epithelial cells (PTs), whereas PAD4 knockout inhibited PANoptosis signalling. PTs expressed mainly RIPK1-PANoptosomes, which executed NET-induced PANoptosis in PTs in renal IRI model mice. Mechanistically, NET-derived double-stranded RNA (dsRNA) promoted PANoptosis in PTs, and PT-expressed TLR3 was responsible for the sensing the extracellular dsRNA. Treating mice with chemical inhibitors of the dsRNA/TLR3 complex suppressed PANoptosis and alleviated tissue injury in renal IRI. Together, the results of this study reveal a mechanism by which the NET-dsRNA-TLR3 axis aggravates PT cell PANoptosis in renal IRI.
Collapse
Affiliation(s)
- Shaoyong Zhuang
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai, 200127, China
| | - Fangzhou Li
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai, 200127, China
- Department of Urology, Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, 201800, China
| | - Liya Wang
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Zilong Lai
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai, 200127, China
| | - Dawei Li
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai, 200127, China
| | - Haoyu Wu
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai, 200127, China
| | - Jiajin Wu
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai, 200127, China
| | - Junwen Qu
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai, 200127, China
| | - Xianyun Zhang
- Department of Urology, The Affiliated Huaian Hospital of Xuzhou Medical University, Jiangsu, 223200, China.
| | - Ming Zhang
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai, 200127, China.
| | - Ruoyang Chen
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai, 200127, China.
| | - Xiaodong Yuan
- Department of Urology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 1630 Dongfang Road, Shanghai, 200127, China.
| |
Collapse
|
41
|
Huang CL, Qu HS, Li AL, Ying CQ, Shao H, Tang YZ, Chen HZ, Tung TH, Zhu JS. Design of a Highly Active Peptide Inhibitor of Farnesyltransferase and Its Protective Effect Against Acute Liver Failure. Drug Des Devel Ther 2025; 19:1909-1926. [PMID: 40098903 PMCID: PMC11912918 DOI: 10.2147/dddt.s505541] [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: 12/11/2024] [Accepted: 02/25/2025] [Indexed: 03/19/2025] Open
Abstract
Purpose Acute liver failure (ALF) is a fatal syndrome associated with massive hepatocyte death. Previous studies have found that Farnesyltransferase (FTase) inhibitors improve disease progression in mouse models of endotoxemia, sepsis, and autoimmune hepatitis. PANoptosis is a novel type of programmed cell death (PCD), including pyroptosis, apoptosis, and necrosis, that plays an important role in ALF. This study was designed and investigated whether the FTase inhibitor PD083176 (d2,d3,d5) could attenuate ALF progression by modulating PANoptosis. Methods Combining the technical tools of computational biology, structural biology and pharmacology, we designed and obtained three high-affinity human FTase inhibitors of PD083176(d2,d3,d5). Then, these FTase inhibitors were investigated by animal experiments by administering PD083176(d2,d3,d5) (10 mg/kg) before modeling with LPS (100 μg/kg)/D-GalN (300 mg/kg) or TAA (800 mg/kg). Results We found that ALF induced by LPS/D-GaIN or TAA were associated with increased farnesylated protein in the liver. PD083176(d2,d3,d5) not only inhibited hepatic farnesylated proteins but also significantly attenuated liver injury and mortality in ALF mice. Importantly, PD083176(d2,d3,d5) treatment effectively inhibited hepatocyte apoptosis (Bax, Bcl-xL and TUNEL cell counts), pyroptosis (Caspase-1 and GSDMD), and necrotic apoptosis (RIPK1 and RIPK3). Conclusion Collectively, these findings demonstrate that PD081376(d2,d3,d5) could alleviate LPS/D-GaIN or TAA-induced ALF by regulating apoptosis, pyroptosis, and necrotizing apoptosis, which might provide a new therapeutic strategy and scalability challenge for ALF.
Collapse
Affiliation(s)
- Chun-Lian Huang
- Department of Infectious Diseases, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, 317000, People’s Republic of China
| | - Hang-Shuai Qu
- Department of Public Laboratory, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, 317000, People’s Republic of China
| | - A-Li Li
- Department of Infectious Diseases, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, 317000, People’s Republic of China
| | - Chen-Qian Ying
- Department of Infectious Diseases, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, 317000, People’s Republic of China
| | - Hui Shao
- Department of Infectious Diseases, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, 317000, People’s Republic of China
| | - Yong-Zhi Tang
- Department of Infectious Diseases, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, 317000, People’s Republic of China
| | - Hua-Zhong Chen
- Department of Infectious Diseases, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, 317000, People’s Republic of China
| | - Tao-Hsin Tung
- Evidence-Based Medicine Center, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, 317000, People’s Republic of China
| | - Jian-Sheng Zhu
- Department of Infectious Diseases, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, Zhejiang, 317000, People’s Republic of China
| |
Collapse
|
42
|
Zhang Y, Lu H, Hou L, Zhang X, Guo T, Wang R, Wang Q, Xing M. GPR120 exacerbates the immune-inflammatory response in chicken liver by mediating acetochlor induced macrophage M1 polarization. JOURNAL OF HAZARDOUS MATERIALS 2025; 485:136928. [PMID: 39709819 DOI: 10.1016/j.jhazmat.2024.136928] [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: 10/21/2024] [Revised: 12/11/2024] [Accepted: 12/16/2024] [Indexed: 12/24/2024]
Abstract
Acetochlor is a widely used and highly effective herbicide. Its overuse poses significant threats to biosecurity and ecological integrity, particularly affecting free-ranging birds. Data on its impact, especially mechanisms of liver toxicity in chickens, are lacking. Thus, we established an animal-cell-animal model to explore intrinsic mechanisms at multiple levels. We found that acetochlor exposure caused liver cell swelling, inflammatory cell accumulation, and lipid deposition. Transcriptomic analyses revealed that differential gene were mainly enriched in hepatic immune, inflammatory, and programmed cell death pathways. We next focused on the gene GPR120, conducting transfection and agonism experiments in LMH, HD11, and co-cultured cells. Acetochlor significantly increased ROS accumulation, activated the NLRP3 inflammasome, and which induced PANoptosis. HD11 cells exhibited M1 polarization with upregulated pro-inflammatory factors. Silencing GPR120 exacerbated cellular damage and immune responses, whereas its agonist, GSK7A, dramatically reduced macrophage M1 polarization and mitigated immune damage to LMH cells. Finally, we returned to animal studies, adding Omega-3-a known GPR120 agonist-to the diet. Omega-3 effectively reversed acetochlor-induced hepatitis and PANoptosis. Given that acetochlor residues pose potential threats to ecosystems and avian health, it is crucial to strengthen residue control, conduct risk assessments, and explore targeted pathways and nutritional supplementation to counteract these negative impacts.
Collapse
Affiliation(s)
- Yue Zhang
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, Heilongjiang 150040, PR China
| | - Hongmin Lu
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, Heilongjiang 150040, PR China
| | - Lulu Hou
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, Heilongjiang 150040, PR China
| | - Xin Zhang
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, Heilongjiang 150040, PR China
| | - Tiantian Guo
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, Heilongjiang 150040, PR China
| | - Ruoqi Wang
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, Heilongjiang 150040, PR China
| | - Qi Wang
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, Heilongjiang 150040, PR China
| | - Mingwei Xing
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin, Heilongjiang 150040, PR China.
| |
Collapse
|
43
|
Zhou H, Zhou M, Liao X, Zhang L, Wei H, Lu Y, Zhang Y, Huang H, Hu Y, Chen T, Lv Z. The Innate Immune Sensor Zbp1 Mediates Central Nervous System Inflammation Induced by Angiostrongylus Cantonensis by Promoting Macrophage Inflammatory Phenotypes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413675. [PMID: 39853924 PMCID: PMC11923990 DOI: 10.1002/advs.202413675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 01/14/2025] [Indexed: 01/26/2025]
Abstract
Angiostrongylus cantonensis (AC) is the leading cause of eosinophilic meningoencephalitis worldwide. The neuroimmune interactions between peripheral and central immune systems in angiostrongyliasis remain unclear. In this study, significant infiltration of eosinophils, myeloid cells, macrophages, neutrophils, and Ly6C monocytes is observed in the brains of AC-infected mice, with macrophages being the most abundant. RNA-seq and SMART-seq analysis of pattern recognition receptor (PRR) and DNA sensor gene sets revealed a marked increase in Z-DNA binding protein 1 (Zbp1) expression in infected mice. Confocal microscopy, RT-qPCR, western blotting, and immunohistochemistry further confirmed that Zbp1 is specifically upregulated in macrophages and microglia. Using Zbp1-knockout mice and flow cytometry, it is found that knockout of Zbp1 enhanced lymphocyte infiltration and natural killer cell cytotoxicity, modulating the immune microenvironment in the central nervous system (CNS) during AC infection. Mechanistically, it is revealed that in macrophage Zbp1 directly binds to receptor-interacting protein 3 (RIP3) to promote its phosphorylation, subsequently facilitating the phosphorylation of mixed lineage kinase domain-like protein (Mlkl). The activated Zbp1-pRIP3-pMlkl axis leads to necroptosis and upregulates pro-inflammatory cytokines including TNF-α, IL-1α, CXCL9, CXCL10 in macrophages, which recruits and activates immune cells. These findings offer new insights into the pathogenic mechanisms of angiostrongyliasis and suggest potential therapeutic strategies.
Collapse
Affiliation(s)
- Hongli Zhou
- Key Laboratory of Tropical Disease Control (Sun Yat‐Sen University)Ministry of EducationGuangzhouGuangdong510080China
- Clinical Medical Research CenterThe Second Affiliated HospitalArmy Medical UniversityChongqing400037China
| | - Minyu Zhou
- Key Laboratory of Tropical Disease Control (Sun Yat‐Sen University)Ministry of EducationGuangzhouGuangdong510080China
- Provincial Engineering Technology Research Center for Biological Vector ControlSun Yat‐sen UniversityGuangzhou510080China
| | - XiPing Liao
- Clinical Medical Research CenterThe Second Affiliated HospitalArmy Medical UniversityChongqing400037China
| | - Liangyu Zhang
- Key Laboratory of Tropical Disease Control (Sun Yat‐Sen University)Ministry of EducationGuangzhouGuangdong510080China
- Provincial Engineering Technology Research Center for Biological Vector ControlSun Yat‐sen UniversityGuangzhou510080China
| | - Hang Wei
- Key Laboratory of Tropical Disease Control (Sun Yat‐Sen University)Ministry of EducationGuangzhouGuangdong510080China
- Provincial Engineering Technology Research Center for Biological Vector ControlSun Yat‐sen UniversityGuangzhou510080China
| | - Yuting Lu
- Key Laboratory of Tropical Disease Control (Sun Yat‐Sen University)Ministry of EducationGuangzhouGuangdong510080China
- Provincial Engineering Technology Research Center for Biological Vector ControlSun Yat‐sen UniversityGuangzhou510080China
| | - Yiqing Zhang
- Key Laboratory of Tropical Disease Control (Sun Yat‐Sen University)Ministry of EducationGuangzhouGuangdong510080China
- Provincial Engineering Technology Research Center for Biological Vector ControlSun Yat‐sen UniversityGuangzhou510080China
| | - Hui Huang
- Key Laboratory of Tropical Disease Control (Sun Yat‐Sen University)Ministry of EducationGuangzhouGuangdong510080China
- Provincial Engineering Technology Research Center for Biological Vector ControlSun Yat‐sen UniversityGuangzhou510080China
| | - Yue Hu
- Key Laboratory of Tropical Disease Control (Sun Yat‐Sen University)Ministry of EducationGuangzhouGuangdong510080China
- Provincial Engineering Technology Research Center for Biological Vector ControlSun Yat‐sen UniversityGuangzhou510080China
| | - Tao Chen
- Department of NeurologyHainan General Hospital,Hainan Affiliated Hospital of Hainan Medical UniversityHaikouHainan570311China
| | - Zhiyue Lv
- Key Laboratory of Tropical Disease Control (Sun Yat‐Sen University)Ministry of EducationGuangzhouGuangdong510080China
- Provincial Engineering Technology Research Center for Biological Vector ControlSun Yat‐sen UniversityGuangzhou510080China
- Department of NeurologyHainan General Hospital,Hainan Affiliated Hospital of Hainan Medical UniversityHaikouHainan570311China
- Key Laboratory of Tropical Translational Medicine of Ministry of EducationHainan Medical UniversityHaikouHainan570216China
| |
Collapse
|
44
|
Kang Y, Yin S, Zhou X, Liu J, Tan X, Zhang C, Lai S, Shao L. Intranasal Zinc Oxide Nanoparticles Induce Neuronal PANoptosis via Microglial Pathway. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2408139. [PMID: 40012250 DOI: 10.1002/smll.202408139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 12/12/2024] [Indexed: 02/28/2025]
Abstract
Recent data have revealed an increased risk of respiratory exposure during the manufacturing process and application of nanomaterials, resulting in an increased incidence of neurodegenerative diseases in the general population. Zinc oxide nanoparticles (ZNPs) are among the most used nanomaterials in biomedical and manufactured consumer products. In this study, neurological dysfunction after intranasal administration of ZNPs is observed, in which the ZNPs enter the brain via the nose-to-brain pathway and accumulate in microglia but not in astrocytes or neurons. By using a coculture system of microglia and neurons, the ZNPs are found that induce microglia-derived oxidative stress injury and lead to neuronal cell PANoptosis. In this context, ZNPs induced the generation of reactive oxygen species (ROS) originating from microglial NADPH oxidase 2 (NOX2), which further induced neuronal membrane lipid peroxidation and increased Ca2+ influx and mitochondrial DNA release. The leaked mitochondrial DNA subsequently initiates PANoptosis of neurons. Importantly, inhibition of microglial NOX2 activation can significantly alleviate brain oxidative injury and rescue neuronal PANoptosis. This study can advance the understanding of the mode of neuronal cell death while underscoring the importance of the interconnections among glial cells and neurons, which is beneficial for informing effective interventions for respiratory exposure to nanoparticles.
Collapse
Affiliation(s)
- Yiyuan Kang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Guangzhou, 510515, China
| | - Suhan Yin
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Xinru Zhou
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Jia Liu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Xiner Tan
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Can Zhang
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Shulin Lai
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
| | - Longquan Shao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, China
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, Guangzhou, 510515, China
| |
Collapse
|
45
|
Su X, Sun Y, Dai A. New insights into pulmonary arterial hypertension: interaction between PANoptosis and perivascular inflammatory responses. Apoptosis 2025:10.1007/s10495-025-02086-0. [PMID: 39979525 DOI: 10.1007/s10495-025-02086-0] [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: 01/24/2025] [Indexed: 02/22/2025]
Abstract
Pulmonary arterial hypertension (PAH) is a heterogeneous disease characterized by various etiologies, with pulmonary vascular remodeling recognized as a main pathological change. Currently, it is widely accepted that vascular remodeling is closely associated with abnormal pulmonary vascular cell death and perivascular inflammation. The simultaneous activation of various pulmonary vascular cell death leads to immune cell adhesion and inflammatory mediator releases; And in turn, the inflammatory response may also trigger cell death and jointly promote the progression of vascular remodeling. Recently, PANoptosis has been identified as a phenomenon that describes the simultaneous activation and interaction of multiple forms of programmed cell death (PCD). Therefore, the relationship between PANoptosis and inflammation in PAH warrants further investigation. This review examines the mechanisms underlying apoptosis, necroptosis, pyroptosis, and inflammatory responses in PAH, with a focus on PANoptosis and its interactions with inflammation. And it aims to elucidate the significance of this emerging form of cell death and inflammation in the pathophysiology of PAH and to explore its potential as a therapeutic target.
Collapse
Affiliation(s)
- Xianli Su
- College of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, People's Republic of China
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China
| | - Yinhui Sun
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, 410208, People's Republic of China
| | - Aiguo Dai
- Hunan Provincial Key Laboratory of Vascular Biology and Translational Medicine, Changsha, Hunan, 410208, People's Republic of China.
- Department of Respiratory Medicine, School of Medicine, Changsha, Hunan, 410021, People's Republic of China.
- Department of Respiratory Medicine, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, 410021, People's Republic of China.
| |
Collapse
|
46
|
Peng J, Tong L, Liang R, Yan H, Jiang X, Dai Y. PANoptosis-Related Optimal Model (PROM): A Novel Prognostic Tool Unveiling Immune Dynamics in Lung Adenocarcinoma. Int J Genomics 2025; 2025:5595391. [PMID: 40008397 PMCID: PMC11858721 DOI: 10.1155/ijog/5595391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 01/16/2025] [Indexed: 02/27/2025] Open
Abstract
Background: PANoptosis, a recently characterized inflammatory programmed cell death modality orchestrated by the PANoptosome complex, integrates molecular mechanisms of pyroptosis, apoptosis, and necroptosis. Although this pathway potentially mediates tumor progression, its role in lung adenocarcinoma (LUAD) remains largely unexplored. Methods: Through comprehensive single-cell transcriptomic profiling, we systematically identified critical PANoptosis-associated gene signatures. Prognostic molecular determinants were subsequently delineated via univariate Cox proportional hazards regression analysis. We constructed a PANoptosis-related optimal model (PROM) through the integration of 10 machine learning algorithms. The model was initially developed using The Cancer Genome Atlas (TCGA)-LUAD cohort and subsequently validated across six independent LUAD cohorts. Model performance was evaluated using mean concordance index. Furthermore, we conducted extensive multiomics analyses to delineate differential pathway activation patterns and immune cell infiltration profiles between PROM-stratified risk subgroups. Results: Cellular populations exhibiting elevated PANoptosis signatures demonstrated enhanced intercellular signaling networks. PROM demonstrated superior prognostic capability across multiple validation cohorts. Receiver operating characteristic curve analyses revealed area under the curve values exceeding 0.7 across all seven cohorts, with several achieving values above 0.8, indicating robust discriminative performance. The model score exhibited significant correlation with immunological parameters. Notably, high PROM scores were associated with attenuated immune responses, suggesting an immunosuppressive tumor microenvironment. Multiomics investigations revealed significant alterations in critical oncogenic pathways and immune landscape between PROM-stratified subgroups. Conclusion: This investigation establishes PROM as a clinically applicable prognostic tool for LUAD risk stratification. Beyond its predictive utility, PROM elucidates PANoptosis-associated immunological and biological mechanisms underlying LUAD progression. These findings provide novel mechanistic insights into LUAD pathogenesis and may inform the development of targeted therapeutic interventions and personalized treatment strategies to optimize patient outcomes.
Collapse
Affiliation(s)
- Jianming Peng
- School of Medicine, Yangzhou Polytechnic College, Yangzhou, China
| | - Leijie Tong
- Department of Immunology, China Medical University, Shenyang, China
| | - Rui Liang
- School of Basic Medical Science, Suzhou Vocational Health College, Suzhou, China
| | - Huisen Yan
- School of Medicine, Yangzhou Polytechnic College, Yangzhou, China
| | - Xiuling Jiang
- School of Medicine, Yangzhou Polytechnic College, Yangzhou, China
| | - Youai Dai
- Laboratory of Organ Transplantation Research Institute, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, China
| |
Collapse
|
47
|
Li Y, Zou H, Ma L, Hu D, Long H, Lin J, Luo Z, Zhou Y, Liao F, Wang X, Meng Y, Wang W, Li G, Zhang Z. Fuzheng Jiedu decoction alleviates H1N1 virus-induced acute lung injury in mice by suppressing the NLRP3 inflammasome activation. JOURNAL OF ETHNOPHARMACOLOGY 2025; 341:119314. [PMID: 39746408 DOI: 10.1016/j.jep.2024.119314] [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: 06/11/2024] [Revised: 12/28/2024] [Accepted: 12/30/2024] [Indexed: 01/04/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Severe influenza, marked by excessive cytokine production, is a major contributor to death in hospitalized individuals. Fuzheng Jiedu decoction (FZJDD), an effective traditional Chinese herbal recipe, has demonstrated promising results in combating the COVID-19 pandemic by reducing mortality and improving Symptoms, and has exhibited anti-inflammatory properties in both clinical trials and laboratory research. Given that pneumonia is a common outcome of SARS-CoV-2 and H1N1 virus infections, we hypothesized that FZJDD may also have therapeutic effects on influenza-related pneumonia and acute lung injury (ALI). AIM OF THE STUDY This research sought to explore the impact and underlying mechanisms of FZJDD on ALI caused by the H1N1 virus in mice. MATERIALS AND METHODS FZJDD was characterized using UHPLC-MS/MS. A mouse model infected with H1N1 virus was used to examine the therapeutic and protective benefits of FZJDD in a living organism, by monitoring body weight fluctuations, lung index, histopathological changes, lung injury scores, and survival rates. Lung tissues underwent haematoxylin-eosin staining, western blotting, qRT-PCR and plaque reduction assay. Blood serum was gathered to assess levels of IL-1β, IL-6, TNF-α through ELISA testing. The impact of FZJDD on the NLRP3 inflammasome was further evaluated in macrophages. RESULTS FZJDD treatment significantly mitigated weight loss, reduced lung index, alleviated histopathological injury, and improved the survival rates in mice with H1N1 virus-induced ALI, demonstrating a protective effect against influenza virus infection. qRT-PCR and Western blot assays revealed that FZJDD treatment ameliorated the hyperinflammatory response caused by the H1N1 virus in lung tissue by suppressing NLRP3 inflammasome activation, without impacting viral replication. In vitro experiments additionally verified that FZJDD treatment can suppress the activation of the NLRP3 inflammasome triggered by the H1N1 virus. CONCLUSION Our findings demonstrate that FZJDD treatment can mitigate ALI caused by H1N1 virus and enhance the survival rate in mice, while it doesn't lower viral titers in the lungs. FZJDD achieves these outcomes by curbing excessive inflammation and blocking the activation of NLRP3 inflammasome.
Collapse
Affiliation(s)
- Yuting Li
- Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Haimei Zou
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Lin Ma
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Dingwen Hu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Haishan Long
- Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Jingnan Lin
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510120, China
| | - Ziqing Luo
- Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Ye Zhou
- Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Feng Liao
- Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Xianyang Wang
- Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Yu Meng
- Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Wenbiao Wang
- Medical Research Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China.
| | - Geng Li
- Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China; Chinese Medicine Guangdong Laboratory, Guangdong, Hengqin, 519031, China; State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou, 510120, China.
| | - Zhongde Zhang
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, 510120, China; Chinese Medicine Guangdong Laboratory, Guangdong, Hengqin, 519031, China; State Key Laboratory of Traditional Chinese Medicine Syndrome, Guangzhou University of Chinese Medicine, Guangzhou, 510120, China.
| |
Collapse
|
48
|
Cai Y, Xiao H, Xue S, Li P, Zhan Z, Lin J, Song Z, Liu J, Xu W, Zhou Q, Qi S, Zhang X, Luo Z. Integrative analysis of immunogenic PANoptosis and experimental validation of cinobufagin-induced activation to enhance glioma immunotherapy. J Exp Clin Cancer Res 2025; 44:35. [PMID: 39901195 PMCID: PMC11789371 DOI: 10.1186/s13046-025-03301-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 01/23/2025] [Indexed: 02/05/2025] Open
Abstract
BACKGROUND Glioma, particularly glioblastoma (GBM), is a highly aggressive tumor with limited responsiveness to immunotherapy. PANoptosis, a form of programmed cell death merging pyroptosis, apoptosis, and necroptosis, plays an important role in reshaping the tumor microenvironment (TME) and enhancing immunotherapy effectiveness. This study investigates PANoptosis dynamics in glioma and explores the therapeutic potential of its activation, particularly through natural compounds such as cinobufagin. METHODS We comprehensively analyzed PANoptosis-related genes (PANoRGs) in multiple glioma cohorts, identifying different PANoptosis patterns and constructing the PANoptosis enrichment score (PANoScore) to evaluate its relationship with patient prognosis and immune activity. Cinobufagin, identified as a PANoptosis activator, was evaluated for its ability to induce PANoptosis and enhance anti-tumor immune responses both in vitro and in vivo GBM models. RESULTS Our findings indicate that high PANoScore gliomas showed increased immune cell infiltration, particularly effector T cells, and enhanced sensitivity to immunotherapies. Cinobufagin effectively induced PANoptosis, leading to increased immunogenic cell death, facilitated tumor-associated microglia/macrophages (TAMs) polarization towards an M1-like phenotype while augmenting CD4+/CD8 + T cell infiltration and activation. Importantly, cinobufagin combined with anti-PD-1 therapy exhibited significant synergistic effects and prolonged survival in GBM models. CONCLUSIONS These findings highlight the therapeutic potential of PANoptosis-targeting agents, such as cinobufagin, in combination with immunotherapy, offering a promising approach to convert "cold" tumors into "hot" ones and improving glioma treatment outcomes.
Collapse
Affiliation(s)
- Yonghua Cai
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Heng Xiao
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Shuaishuai Xue
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Peng Li
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Zhengming Zhan
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Jie Lin
- Department of Neurosurgery, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, Guangzhou, 510623, China
| | - Zibin Song
- Neurosurgery Center, Department of Functional Neurosurgery, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jun Liu
- Department of Neurosurgery, the 2nd affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Wei Xu
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Qixiong Zhou
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Songtao Qi
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| | - Xi'an Zhang
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| | - Ziyi Luo
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China.
- Department of Neurosurgery, Guangzhou Women and Children's Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou Medical University, Guangzhou, 510623, China.
| |
Collapse
|
49
|
Li F, Zhang Q, Rong Y, Xiang S, Wang J. TAT-N24 enhances retinal ganglion cell survival by suppressing ZBP1-PANoptosome-mediated PANoptosis in an acute glaucoma mouse model. Exp Eye Res 2025; 251:110244. [PMID: 39832635 DOI: 10.1016/j.exer.2025.110244] [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: 08/21/2024] [Revised: 12/18/2024] [Accepted: 01/17/2025] [Indexed: 01/22/2025]
Abstract
The abrupt and substantial elevation of intraocular pressure (IOP) in acute glaucoma induces retinal ischemia/reperfusion (I/R) injury, resulting in progressive retinal ganglion cell (RGC) death and irreversible visual impairment. PANoptosis, a form of regulated cell death consisting of pyroptosis, apoptosis and necroptosis, is reported to be involved in high IOP-induced RGC death. However, the precise mechanisms of RGC death remain unclear, and neuroinflammation is considered to play a vital role. TAT-N24, a synthetic inhibitor targeting the p55 regulatory subunit of phosphatidylinositol 3-kinase (p55PIK) signaling, demonstrates anti-inflammatory effect in uveitis and may have certain neuroprotective effects. Therefore, we investigated whether TAT-N24 could shield RGCs from immunoinflammatory damage in an acute glaucoma mouse model and explored the potential mechanism associated with PANoptosis. A mouse model of acute ocular hypertension (AOH) was established. Intravitreal injection of TAT-N24 was conducted to evaluate its impact on RGC death. The expression levels of key components in PANoptosis were analyzed using RT-qPCR and Western blotting. Immunohistochemistry and immunofluorescence staining on eyeball sections were employed to assess the expression of p55PIK, Brn3a, and ionized calcium binding adaptor molecule 1 (Iba1). Retinal structure was examined by H&E staining, while cell apoptosis was evaluated by TdT-mediated dUTP nick end labeling (TUNEL). The results showed that intravitreal injection of TAT-N24 effectively alleviated RGC death and retinal damage induced by AOH injury. The key components in PANoptosis were markedly upregulated after AOH injury, while these components were significantly inhibited after TAT-N24 treatment. Moreover, the expression levels of Z-DNA-binding protein 1 (ZBP1)-PANoptosome (ZBP1, RIPK1, RIPK3, and Caspase-8), NLR family pyrin domain-containing protein 3 (NLRP3), and NLR family CARD domain-containing protein 4 (NLRC4) inflammasomes were notably elevated after AOH injury, which was significantly suppressed by TAT-N24. In conclusion, PANoptosis was involved in AOH-induced RGC death and retinal damage. TAT-N24 exhibited an anti-PANoptotic effect, protecting RGCs by inhibiting ZBP1-PANoptosome as well as NLRP3 and NLRC4 inflammasomes after AOH injury.
Collapse
Affiliation(s)
- Fei Li
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qiuxiang Zhang
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yan Rong
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Sifei Xiang
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Junming Wang
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
50
|
Jiajia D, Wen Y, Enyan J, Xiaojian Z, Zhen F, Jia Z, Jikai W, Xiaoxin Y, Aihua L, Fangen K, Fei L. PGAM5 promotes RIPK1-PANoptosome activity by phosphorylating and activating RIPK1 to mediate PANoptosis after subarachnoid hemorrhage in rats. Exp Neurol 2025; 384:115072. [PMID: 39603487 DOI: 10.1016/j.expneurol.2024.115072] [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: 08/24/2024] [Revised: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 11/29/2024]
Abstract
Neuronal death plays a crucial role in early brain injury after subarachnoid hemorrhage (SAH). PANoptosis is a programmed form of cell death regulated by the PANoptosome, which possesses key characteristics of pyroptosis, apoptosis and necroptosis. Phosphoglycerate mutase family member 5 (PGAM5) has specific phosphatase activity that phosphorylates or dephosphorylates serine and threonine residues on bound proteins such as receptor-interacting protein kinase 1 (RIPK1), which are involved in programmed cell death. This study aimed to explore whether PANoptosis occurs after subarachnoid hemorrhage and to investigate the role of PGAM5 in early brain injury after SAH. A monofilament perforation SAH model in Sprague-Dawley rats was established, and PGAM5 siRNA (siPGAM5) was administered via intracerebroventricular injection 48 h before SAH modeling. The efficacy of siPGAM5 treatment was assessed via neurological scoring, and the impact of siPGAM5 on PANoptosis was evaluated via Western blotting, TUNEL staining and ELISA. To investigate its potential mechanism, the RIPK1 activator birinapant was administered intraperitoneally 0.5 h after SAH. The role of RIPK in PGAM5-mediated PANoptosis was evaluated by Western blotting and coimmunoprecipitation. Our findings indicate that PANoptosis occurs in neurons after SAH and that reducing PGAM5 in the cytosol after SAH can reduce PANoptosis and enhance the short-term and long-term neurological functions of SAH rats. Mechanistically, we discovered that PGAM5 can directly bind to and phosphorylate and activate RIPK1 (ser 166), triggering the assembly of the RIPK1-PANoptosome complex. In conclusion, our study revealed that the increased PGAM5 in the mitochondria-free cytosol after SAH can bind to and activate RIPK1 (ser 166), driving the assembly of the RIPK1-PANoptosome and mediating PANoptosis after SAH. PGAM5 and PANoptosis might be novel therapeutic targets for SAH.
Collapse
Affiliation(s)
- Duan Jiajia
- Department of Neurosurgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China; Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Yuan Wen
- Department of Neurosurgery, Zhuzhou Central Hospital, Zhuzhou, Hunan 412000, China
| | - Jiang Enyan
- Department of Neurosurgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China; Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Zhang Xiaojian
- Department of Neurosurgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China; Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Fang Zhen
- Department of Neurosurgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China; Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Zeng Jia
- Department of Neurosurgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China; Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Wang Jikai
- Department of Neurosurgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China; Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Yan Xiaoxin
- Department of Anatomy and Neurobiology, Xiangya Medicine School, Central South University, Changsha, Hunan, China
| | - Liu Aihua
- Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Kong Fangen
- Department of Neurosurgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China; Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China.
| | - Liu Fei
- Department of Neurosurgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China; Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China.
| |
Collapse
|