1
|
Hu C, Yuan F, Wu Y, Xiao S, Xu Y, Peng X, He L. Disruption of the caspase-1/IL-1β axis alleviates myocardial Ischemia/Reperfusion injury via improvement of mitochondrial homeostasis and reduction of Pyroptosis. Clin Exp Hypertens 2025; 47:2506619. [PMID: 40373207 DOI: 10.1080/10641963.2025.2506619] [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: 03/06/2025] [Revised: 04/28/2025] [Accepted: 05/09/2025] [Indexed: 05/17/2025]
Abstract
BACKGROUND Pyroptosis is a novel kind of programmed cell death and Caspase-1 plays key roles in driving pyroptosis. The current study aims to elucidate the molecular mechanism affecting cardiomyocyte pyroptosis in myocardial ischemia/reperfusion (I/R) injury, both in vivo and in vitro. METHODS A murine model of myocardial I/R injury was established and then treated with lentivirus-mediated shRNA targeting Caspase-1 to evaluate the effect of Caspase-1 on myocardial I/R injury. Further, Caspase-1 was silenced in the cardiomyocytes following hypoxia-reoxygenation (H/R) to detect the function of Caspase-1 in mitochondrial homeostasis and cardiomyocyte pyroptosis. RESULTS Knockdown of Caspase-1 inhibited the secretion of interleukin-1 beta (IL-1β), improved cardiac dysfunction and decreased pyroptosis in vivo. The cardio-protective effect was verified in the H/R-induced cardiomyocyte model. Recombinant IL-1β protein reversed the inhibitory effect of Caspase-1 knockdown on pyroptosis. CONCLUSION Overall, activating the Caspase-1/IL-1β axis by myocardial I/R injury causes mitochondrial homeostasis imbalance, pyroptosis, and the consequent cardiomyocyte injury.
Collapse
Affiliation(s)
- ChenKai Hu
- Department of Cardiology, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - FengXia Yuan
- Department of Pharmacy, the Second Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi Province, China
| | - YingXing Wu
- Department of Cardiology, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Shan Xiao
- Department of Cardiology, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Yuan Xu
- Medical Big Data Research Center, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Xiang Peng
- Information Department, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Lei He
- Department of Cardiology, the Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| |
Collapse
|
2
|
Luo Y, Xu D, Yu C. Research progress on sepsis-associated encephalopathy by inhibiting pyroptosis. Gene 2025; 961:149560. [PMID: 40355013 DOI: 10.1016/j.gene.2025.149560] [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: 02/15/2025] [Revised: 04/28/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025]
Abstract
Sepsis is a life-threatening condition characterized by multiple organ dysfunction syndrome resulted from dysregulated host responses to infection. Sepsis-associated encephalopathy (SAE) is one of the most common symptoms of acute-phase sepsis, with nearly 70 % of patients with sepsis ultimately developing SAE. Pyroptosis represents a type of cell death that is initiated by inflammation. This cell death type is associated with various infectious and noninfectious diseases. The gasdermin family proteins are crucial cell death executors and critical components in regulating the canonical pyroptosis pathway in microglia. In this review, we summarize the inhibitory effects of several drugs and genes on the pyroptosis pathway. Our findings suggest that several drugs (puerarin, VX765, HC067047, dexpramipexole, and Danhong injection), erbin gene, and TRIM45 knockdown improve SAE by suppressing the canonical pathway of NLRP3/caspase-1/gasdermin D-mediated pyroptosis. Therefore, they have significant importance in terms of brain protection. Moreover, we review the relevant literature published in recent years and summarize the research status and development prospects in this field to provide a basis for subsequent related research.
Collapse
Affiliation(s)
- Yanhua Luo
- Department of Yanbian University Hospital, Yanji, Jilin 133000, People's Republic of China
| | - Dahai Xu
- Department of Emergency Medicine, The First Hospital of Jilin University, Changchun Jilin 130000, People's Republic of China
| | - Chenglin Yu
- Department of Emergency Medicine, Yanbian University Hospital, Yanji, Jilin 133000, People's Republic of China.
| |
Collapse
|
3
|
Cai J, Zhao J, Peng R, Yu H, He Y, Zhou Q, Wang Y, Xie P. NLRP3 in the dorsal raphe nucleus manipulates the depressive-like behaviors. Brain Res Bull 2025; 227:111405. [PMID: 40447162 DOI: 10.1016/j.brainresbull.2025.111405] [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: 03/15/2025] [Revised: 05/24/2025] [Accepted: 05/26/2025] [Indexed: 06/02/2025]
Abstract
Major depressive disorder is one of the most common psychiatric disorders, and the Nod-like receptor family pyrin domain containing 3 (NLRP3) inflammasome plays an important role in depression. Dorsal raphe nucleus (DRN), as the main origin of producing serotonin in the brain, is an important functional brain region in depressive disorders. However, the relationship between NLRP3 in the DRN and depression has not been clarified in previous studies. So, we focus on demonstrating the role of NLRP3 expressed in DRN in depression. In this study, the male C57BL/6 J mice were exposed to chronic unpredictable mild stimulation and the expression and cellular localization of NLRP3 in DRN were analyzed. Subsequently, the mice were treated with the NLRP3 inhibitor MCC950 to inhibit NLRP3 inflammasome, and the expression of NLRP3 was knocked down in certain cells within the DRN of NLRP3fl/fl mice to investigate the role of NLRP3 in regulating depressive phenotype. Compared with the control group, the expression of NLRP3 in DRN of CUMS group was significantly increased, especially in the microglia and neuron. Furthermore, treatment with the NLRP3 inhibitor induced a significant antidepressant effect, and the depressive phenotype of NLRP3fl/fl mice was rescued after knocking down NLRP3 in the microglia or neuron. In addition, the expression levels of related molecules in the NLRP3 inflammasome pathway were significantly higher in the CUMS group compared to the control group. These results illustrated that NLRP3 played an important role in regulating depressive phenotype in DRN, and suggested a new therapy target for depression.
Collapse
Affiliation(s)
- Junchao Cai
- Department of Neurology, NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Jiarong Zhao
- Department of Neurology, NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Rui Peng
- Department of Neurology, NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Heming Yu
- Department of Neurology, NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yong He
- Department of Neurology, NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Qigang Zhou
- State Key Laboratory of Reproductive Medicine, Department of Clinic Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Yue Wang
- Department of Neurology, NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| | - Peng Xie
- Department of Neurology, NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Neurology, Yongchuan Hospital of Chongqing Medical University, Chongqing 402460, China; Chongqing Key Laboratory of Neurobiology, Chongqing 400016, China; Chongqing Institute for Brain and Intelligence, Chongqing 401336, China.
| |
Collapse
|
4
|
Xiao S, Shen Y, Zhang M, Liu X, Cai T, Wang F. VacA promotes pyroptosis via TNFAIP3/TRAF1 signaling to induce onset of atrophic gastritis. Microbiol Res 2025; 296:128142. [PMID: 40138873 DOI: 10.1016/j.micres.2025.128142] [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: 02/09/2025] [Revised: 03/10/2025] [Accepted: 03/13/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Atrophic gastritis (AG) is a chronic inflammation where gastric glandular cells are replaced by intestinal-type epithelium. Gastric epithelial cell loss is often linked to multiple cell death signaling pathways. While Helicobacter pylori (H. pylori) infection is the main cause of AG, its role in inducing cell death goes beyond apoptosis and autophagy. Pyroptosis could promote development of inflammation related cancers, but its involvement in H. pylori-induced malignant transformation remains unclear. METHODS The enrichment of pyroptosis signaling across pathological stages was assessed using immunohistochemistry and bioinformatic analysis. Gastric epithelial cells were co-cultured with VacA recombinant protein or VacA+H. pylori to investigate the role of VacA in pyroptosis, and its downstream targets. TNFAIP3 or TRAF1 was silenced/overexpressed in gastric epithelial cells to explore their impact on pyroptosis. Finally, the interaction between TNFAIP3 and TRAF1 was examined using Western Blot, immunofluorescence, co-immunoprecipitation and ubiquitin assays. RESULTS Expression of pyroptosis components and pyroptosis enrichment score were upregulated in AG and gastric cancer tissues compared to normal or non-atrophic gastritis tissues. Upon incubation with VacA recombinant protein or VacA+H. pylori, pyroptosis and TNFAIP3/TRAF1 were elevated in gastric epithelial cells. TRAF1 promoted expression of downstream pyroptosis components and release of IL-1β/IL18. TRAF1 ablation could reverse pyroptosis activation caused by VacA. Finally, we proved TNFAIP3 as deubiquitinating enzyme to increase TRAF1 stability, further inducing pyroptosis. CONCLUSIONS The VacA/TNFAIP3/TRAF1 signaling cascade facilitates pyroptosis in H. pylori- infected tissue. Overactivation of Pyroptosis caused the atrophy-like morphological changes of gastric epithelium, further inducing sustainable malignant transformation.
Collapse
Affiliation(s)
- Shilang Xiao
- Department of Gastroenterology, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Changsha, Hunan 410013, China; Hunan Key Laboratory of Non-Resolving Inflammation and Cancer, Changsha, China
| | - Yicun Shen
- Department of Gastroenterology, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Changsha, Hunan 410013, China; Hunan Key Laboratory of Non-Resolving Inflammation and Cancer, Changsha, China
| | - Minglin Zhang
- Department of Gastroenterology, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Changsha, Hunan 410013, China; Hunan Key Laboratory of Non-Resolving Inflammation and Cancer, Changsha, China
| | - Xiaoming Liu
- Department of Gastroenterology, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Changsha, Hunan 410013, China; Hunan Key Laboratory of Non-Resolving Inflammation and Cancer, Changsha, China.
| | - Ting Cai
- Department of gastroenterology, Hunan provincial people's hospital, the first affiliated hospital of Hunan Normal University, 61 Jiefang Road, Changsha, Hunan 410005, China.
| | - Fen Wang
- Department of Gastroenterology, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Changsha, Hunan 410013, China; Hunan Key Laboratory of Non-Resolving Inflammation and Cancer, Changsha, China.
| |
Collapse
|
5
|
Tang K, Ye T, He Y, Ba X, Xia D, Peng E, Chen Z, Ye Z, Yang X. Ferroptosis, necroptosis, and pyroptosis in calcium oxalate crystal-induced kidney injury. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167791. [PMID: 40086520 DOI: 10.1016/j.bbadis.2025.167791] [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: 11/30/2024] [Revised: 01/24/2025] [Accepted: 03/04/2025] [Indexed: 03/16/2025]
Abstract
Kidney stones represent a highly prevalent urological disorder worldwide, with high incidence and recurrence rates. Calcium oxalate (CaOx) crystal-induced kidney injury serves as the foundational mechanism for the formation and progression of CaOx stones. Regulated cell death (RCD) such as ferroptosis, necroptosis, and pyroptosis are essential in the pathophysiological process of kidney injury. Ferroptosis, a newly discovered RCD, is characterized by its reliance on iron-mediated lipid peroxidation. Necroptosis, a widely studied programmed necrosis, initiates with a necrotic phenotype that resembles apoptosis in appearance. Pyroptosis, a type of RCD that involves the gasdermin protein, is accompanied by inflammation and immune response. In recent years, increasing amounts of evidence has demonstrated that ferroptosis, necroptosis, and pyroptosis are significant pathophysiological processes involved in CaOx crystal-induced kidney injury. Herein, we summed up the roles of ferroptosis, necroptosis, and pyroptosis in CaOx crystal-induced kidney injury. Furthermore, we delved into the curative potential of ferroptosis, necroptosis, and pyroptosis in CaOx crystal-induced kidney injury.
Collapse
Affiliation(s)
- Kun Tang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Ye
- Department of Geriatric Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu He
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaozhuo Ba
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ding Xia
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ejun Peng
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiqiang Chen
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhangqun Ye
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoqi Yang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
6
|
Zhang Y, Liu T, Guo H, Shi J, Yang J, Fu S, Pan X, Li F, Zhang H, Zhang D, Yang H, Zheng L, Shi M, Zhou W. Ex vivo lung perfusion with GLP-1R agonist mitigates ischemia/reperfusion injury through pyroptosis modulation in lung transplantation- an experimental study. Int J Surg 2025; 111:3781-3797. [PMID: 40387728 PMCID: PMC12165588 DOI: 10.1097/js9.0000000000002438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 04/14/2025] [Indexed: 05/20/2025]
Abstract
BACKGROUND Ischemia/reperfusion injury (IRI) presents a significant hurdle in lung transplantation. Our previous research showed that the glucagon-like peptide-1 receptor (GLP-1R) agonist liraglutide (Lir) improves lipopolysaccharide-induced acute lung injury in murine models. This study aims to further investigate the lung-protective mechanisms of GLP-1R agonist. METHODS An in vitro hypoxia/reoxygenation (H/R) model with BEAS-2B cells and an in vivo donation after cardiac death (DCD) rat lung transplant model were utilized. Lir was administered using an ex vivo lung perfusion (EVLP) system. Lung function, injury, and pyroptosis mechanisms were assessed. Validation experiments included quantitative reverse transcription PCR, immunoblot analysis, activity assays and proteomic analysis, among others, to evaluate how GLP-1R agonist protect lungs from IRI by modulating pyroptosis, thereby improving lung function and reducing injury. RESULTS Perfusion of the donor lung with Lir using EVLP improved the function of DCD lungs and mitigated IRI. Bioinformatics analysis and validation experiments provided evidence of increased expression of NOD-like receptors signals and pyroptosis in lung transplantation IRI, which was suppressed by Lir treatment. Further investigations revealed that the thioredoxin-binding protein (TXNIP) played a crucial regulatory role in the pyroptosis of IRI, with the NOD-like receptor family pyrin domain-containing 3 (NLRP3) emerging as a key target. In addition, this study found that Lir promotes GLP-1R-dependent TXNIP ubiquitination and modulates TXNIP mRNA stability via the GLP-1R/miR-17 axis. CONCLUSION This study demonstrates, for the first time, that a novel EVLP-based drug delivery approach using GLP-1R agonist can protect lungs from IRI by modulating pyroptosis, thereby improving lung function and reducing injury. The research uncovers a previously unknown mechanism where GLP-1R agonist modulates the protein TXNIP through GLP-1R/miR-17 signaling. These insights underscore the potential of GLP-1R agonists as targeted therapies for primary graft dysfunction in lung transplant recipients, opening new avenues for clinical interventions to improve transplant outcomes.
Collapse
Affiliation(s)
- Yufei Zhang
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tao Liu
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
- Department of Oncology, Huashan Hospital, Fudan University, Shanghai, China
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences and Food Engineering, Liaocheng University, Liaocheng, China
| | - Huaizu Guo
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
- State Key Laboratory of Macromolecular Drugs and Large-scale Preparation, School of Pharmaceutical Sciences and Food Engineering, Liaocheng University, Liaocheng, China
| | - Jianxin Shi
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Yang
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shijie Fu
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xufeng Pan
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng Li
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hai Zhang
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dawei Zhang
- Engineering Research Center of Optical Instrument and System, The Ministry of Education, Shanghai Key Laboratory of Modern Optical System, University of Shanghai for Science and Technology, Shanghai, China
| | - Hong Yang
- The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Lulu Zheng
- Engineering Research Center of Optical Instrument and System, The Ministry of Education, Shanghai Key Laboratory of Modern Optical System, University of Shanghai for Science and Technology, Shanghai, China
| | - Meng Shi
- Department of Thoracic and Cardiovascular Surgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Wenyong Zhou
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
7
|
Chang R, Sun J, Leng J, Wang Z, Mu S, Li Y, Wang J, Song L. A new type of Caspase-1 upon recognizing bacteria inhibits GSDME-dependent histone modification and NF-κB signaling. Commun Biol 2025; 8:827. [PMID: 40442231 PMCID: PMC12122919 DOI: 10.1038/s42003-025-08290-7] [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: 12/16/2024] [Accepted: 05/23/2025] [Indexed: 06/02/2025] Open
Abstract
In the present study, a new type of Caspase-1 homolog is identified from Crassostrea gigas (defined as CgCas1-2D). It is composed of 2×DSRM-CASc domain and has closer evolutionary relationship with mammalian Caspase-1s. The mRNA expressions of CgCas1-2D increase significantly after Vibrio splendidus or LPS stimulation. Recombinant CgCas1-2D and its 2×DSRM and CASc domains all bind various PAMPs and bacteria. rCgCas1-2D shows the highest binding activity to human Caspase-1 substrate. Upon recognizing bacteria, CgCas1-2D co-localizes and interacts with CgGSDME, while it has no cleavage activity to CgGSDME. CgCas1-2D inhibits the histone methylation and acetylation levels and CgNF-κB/Rel nuclear translocation mediated by CgGSDME. In addition, CgCas1-2D suppresses the mRNA expression levels of cytokines mediated by GSDME-NF-κB/Rel axis. The results demonstrate that a new type of anti-inflammatory Caspase-1 identified from oyster upon recognizing various bacteria interacts with GSDME to inhibit the histone modification and NF-κB signaling to suppress the inflammation.
Collapse
Affiliation(s)
- Renle Chang
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, China
- Liaoning Key Laboratory of Marine Animal Immunology & Disease Control, Dalian Ocean University, Dalian, China
- Dalian Key Laboratory of Aquatic Animal Diseases Prevention and Control, Dalian Ocean University, Dalian, China
| | - Jiejie Sun
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, China.
- Liaoning Key Laboratory of Marine Animal Immunology & Disease Control, Dalian Ocean University, Dalian, China.
- Dalian Key Laboratory of Aquatic Animal Diseases Prevention and Control, Dalian Ocean University, Dalian, China.
| | - Jinyuan Leng
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, China
- Liaoning Key Laboratory of Marine Animal Immunology & Disease Control, Dalian Ocean University, Dalian, China
- Dalian Key Laboratory of Aquatic Animal Diseases Prevention and Control, Dalian Ocean University, Dalian, China
| | - Zihan Wang
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, China
- Liaoning Key Laboratory of Marine Animal Immunology & Disease Control, Dalian Ocean University, Dalian, China
- Dalian Key Laboratory of Aquatic Animal Diseases Prevention and Control, Dalian Ocean University, Dalian, China
| | - Shuyi Mu
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, China
- Liaoning Key Laboratory of Marine Animal Immunology & Disease Control, Dalian Ocean University, Dalian, China
- Dalian Key Laboratory of Aquatic Animal Diseases Prevention and Control, Dalian Ocean University, Dalian, China
| | - Yinan Li
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, China
- Liaoning Key Laboratory of Marine Animal Immunology & Disease Control, Dalian Ocean University, Dalian, China
- Dalian Key Laboratory of Aquatic Animal Diseases Prevention and Control, Dalian Ocean University, Dalian, China
| | - Jie Wang
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, China
- Liaoning Key Laboratory of Marine Animal Immunology & Disease Control, Dalian Ocean University, Dalian, China
- Dalian Key Laboratory of Aquatic Animal Diseases Prevention and Control, Dalian Ocean University, Dalian, China
| | - Linsheng Song
- Liaoning Key Laboratory of Marine Animal Immunology, Dalian Ocean University, Dalian, China.
- Liaoning Key Laboratory of Marine Animal Immunology & Disease Control, Dalian Ocean University, Dalian, China.
- Dalian Key Laboratory of Aquatic Animal Diseases Prevention and Control, Dalian Ocean University, Dalian, China.
| |
Collapse
|
8
|
Yang X, Xu C, Zeng Y, Wang C, Gao Y, Ding J, Chen S, Pan Y, Zhang X, Mao Z, Shi S. Pyroptosis-Inducing Platinum(IV) Prodrugs via GSDME Pathway for Chemoimmunotherapy and Metastasis Inhibition in Triple-Negative Breast Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e05567. [PMID: 40432601 DOI: 10.1002/advs.202505567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2025] [Revised: 04/29/2025] [Indexed: 05/29/2025]
Abstract
Pyroptosis has attracted significant attention for its role in cancer chemotherapy and immunotherapy. However, few drugs have been reported to induce pyroptosis via the Caspase-3/gasdermin E (GSDME) pathway. Herein, three novel PtIV prodrugs, MRP, DRP, and HRP are rationally designed by conjugating DNA methyltransferase (DNMT) inhibitor (RG108) and/or histone deacetylase (HDAC) inhibitor (PhB) to the PtIV center. These prodrugs can be easily reduced to cisplatin (CDDP) due to the high glutathione (GSH) levels in tumors, liberating the coordinated ligands. Released RG108 reactivates the GSDME gene and reduces pyroptosis in low GSDME-expressing tumor cells. Meanwhile, PhB-induced chromatin loosening enhances CDDP-DNA binding, which not only increases Caspase-3 expression, but also upregulates GSDME. HRP demonstrates superior ability to suppress tumor growth and metastasis while reducing systemic toxicity compared with CDDP. By reactivating GSDME and loosening chromatin, HRP effectively boosts tumor cell pyroptosis and exhibits the most pronounced anticancer performance. These findings highlight HRP's potential as a therapeutic agent for triple-negative breast cancer (TNBC) and offer innovative strategies for combining chemotherapy with immunotherapy. To the best of current knowledge, this is the first report of platinum complexes inducing pyroptosis via the Caspase-3/GSDME pathway in low GSDME-expressing tumor cells.
Collapse
Affiliation(s)
- Xinda Yang
- School of Chemical Science and Engineering, Department of Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Tongji University, Shanghai, 200092, P. R. China
| | - Chuansheng Xu
- School of Chemical Science and Engineering, Department of Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Tongji University, Shanghai, 200092, P. R. China
| | - Youliang Zeng
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, Guangdong Basic Research Center of Excellence for Functional Molecular Engineering, GBRCE for Functional Molecular Engineering, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510275, P. R. China
| | - Chunhui Wang
- School of Chemical Science and Engineering, Department of Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Tongji University, Shanghai, 200092, P. R. China
| | - Yan Gao
- School of Chemical Science and Engineering, Department of Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Tongji University, Shanghai, 200092, P. R. China
| | - Jie Ding
- School of Chemical Science and Engineering, Department of Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Tongji University, Shanghai, 200092, P. R. China
| | - Sirui Chen
- School of Chemical Science and Engineering, Department of Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Tongji University, Shanghai, 200092, P. R. China
| | - Yuheng Pan
- School of Chemical Science and Engineering, Department of Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Tongji University, Shanghai, 200092, P. R. China
| | - Xin Zhang
- School of Chemical Science and Engineering, Department of Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Tongji University, Shanghai, 200092, P. R. China
| | - Zongwan Mao
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, Guangdong Basic Research Center of Excellence for Functional Molecular Engineering, GBRCE for Functional Molecular Engineering, School of Chemistry, Sun Yat-Sen University, Guangzhou, 510275, P. R. China
| | - Shuo Shi
- School of Chemical Science and Engineering, Department of Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Tongji University, Shanghai, 200092, P. R. China
| |
Collapse
|
9
|
Huang L, Zhang F, Wang Y, Wu J, Wang R, Wei S, Li X, Xu N, Wang Y, Li Y. Functional metabolomics combined with network pharmacology reveals the mechanism of alleviating rheumatoid arthritis with Yiyi Fuzi powder. JOURNAL OF ETHNOPHARMACOLOGY 2025; 348:119842. [PMID: 40268109 DOI: 10.1016/j.jep.2025.119842] [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: 02/12/2025] [Revised: 04/07/2025] [Accepted: 04/18/2025] [Indexed: 04/25/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Yiyi Fuzi powder (YYFZ) is a composite formulation consisting of Fuzi and Coix lacryma-jobi seeds. The synergistic application of these exhibits notable anti-inflammatory properties, playing a crucial role in the management of rheumatoid arthritis (RA). However, the therapeutic advantages and potential mechanism of YYFZ in the treatment of RA are still unclear. AIM OF THE STUDY The purpose of this study is to find functional metabolites by metabolomics technology, and to investigate the mechanism of functional metabolites mediating RA inflammation on the basis of collagen-induced arthritis rat fibroblast-like synovial cells (CIA-FLS) model, and to explore the pharmacodynamic material basis of YYFZ. MATERIALS AND METHODS Utilizing untargeted metabolomics in conjunction with UPLC-Q-TOF/MS and GC-MS, we identified potential functional metabolites of YYFZ. In vitro experiments were conducted to determine pyroptosis-related proteins via Western blot, q-PCR and immunofluorescence, thereby exploring functional metabolic pathways. Subsequently, network pharmacology and molecular docking techniques were employed to evaluate the mode of action and mechanisms of "effective components-key targets", elucidating the active components of YYFZ. RESULTS Using untargeted metabolomics, 18 differential metabolites were identified, with palmitic acid (PA) showing high correlation as a potential functional metabolite. MTT experiments revealed that 300 μM PA inhibited CIA-FLS by 50%. Further analysis through in vitro experiments indicated that PA promotes inflammatory factor expression via NLRP3/Caspase-1/GSDMD-N/IL-1β mediated pyroptosis. Network pharmacology and molecular docking of 26 in vitro YYFZ components identified benzoylaconine (BAC), benzoylmesaconine (BMA) and benzoylhypacoitine (BHA) as potential active components. In vitro experiments revealed that these components reduce RA inflammation by targeting pyroptosis. CONCLUSION PA, a functional metabolite, can promote RA inflammatory factors by inducing pyroptosis of NLRP3/Caspase1/GSDMD-N/IL-1β. BAC, BMA and BHA derived from YYFZ have demonstrated efficacy in mitigating the inflammatory damage induced by the functional metabolite PA, suggesting their potential as therapeutic agents for RA. These findings offer valuable insights for the development of targeted therapies for RA and underscore the clinical applicability of YYFZ.
Collapse
Affiliation(s)
- Liping Huang
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Fangfang Zhang
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yuyu Wang
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Junke Wu
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Rui Wang
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Shuang Wei
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Xinyu Li
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Nanjian Xu
- Department of Spine Surgery, No.6 Hospital in Ningbo, Ningbo city, 315040, China.
| | - Yuming Wang
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Yubo Li
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
10
|
Padhy DS, Vesmaker K, Banerjee S. Neuroprotective potential of tranilast in streptozotocin-induced sporadic Alzheimer's disease model targeting TXNIP-NLRP3 inflammasome pathway. Int Immunopharmacol 2025; 156:114691. [PMID: 40273674 DOI: 10.1016/j.intimp.2025.114691] [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: 02/06/2025] [Revised: 04/10/2025] [Accepted: 04/16/2025] [Indexed: 04/26/2025]
Abstract
Sporadic Alzheimer's disease (sAD) is a progressive neurodegenerative disorder characterised by oxidative stress, neuroinflammation, mitochondrial dysfunction and cerebral insulin resistance. Even though approximately 95 % of AD cases are reported as sporadic, the exact pathogenesis remains sparse. Tranilast, an analogue of tryptophan metabolite, was initially endowed as an anti-allergic agent and used in multiple inflammatory ailments. Still, the molecular mechanisms targeting sAD are yet to be investigated. In the present study, we investigated the neuroprotective potential of tranilast by performing biochemical, molecular and histopathological assessments using both in vivo and in vitro experimental sAD models. Streptozotocin (STZ; 3 mg/kg) was bilaterally injected on day 1 and 3 through the intracerebroventricular (ICV) route to Sprague Dawley rats for the in vivo model induction. Spontaneous alternation test, novel object recognition test, and passive avoidance test were performed to assess the altered behavioural patterns in animals. Furthermore, human neuroblastoma cells (SHSY5Y) were exposed to STZ (1 mM) and tranilast for 24 h to validate the in vivo results. Three weeks of tranilast (30 and 100 mg/kg, p.o.) treatment improved neurobehavioural anomalies in ICV-STZ-treated rats by halting neuroinflammation and NLRP3 inflammasome activation caused by enhanced reactive oxygen species (ROS) and thioredoxin interaction protein (TXNIP) overexpression. The phosphorylated tau (p-tau S416) level was also increased in the ICV-STZ rat's hippocampus and reversed upon tranilast treatment. A high dose of tranilast (100 mg/kg) treatment sensitised hippocampal insulin signalling in ICV-STZ-treated rats. Furthermore, in cell culture studies, 24-h tranilast (30 and 100 μM) treatment reduced the mitochondrial ROS production and attenuated inflammasome activation in STZ-treated SHSY5Y cells. In summary, the findings of the study proclaim the neuroprotective potential of tranilast in STZ induced model of sAD by modulating the TXNIP-NLRP3 inflammasome pathway.
Collapse
Affiliation(s)
- Dibya Sundar Padhy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) - Kolkata, West Bengal, India
| | - Kushal Vesmaker
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) - Kolkata, West Bengal, India
| | - Sugato Banerjee
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER) - Kolkata, West Bengal, India.
| |
Collapse
|
11
|
Zhang X, Deng Z, Zhang X, Xu Q, Liu L, Yang D, Guo Z. Inhibiting cathepsin B alleviates acute lung injury caused by sepsis through suppression of pyroptosis in lung epithelial cells. Eur J Med Res 2025; 30:403. [PMID: 40394663 PMCID: PMC12090420 DOI: 10.1186/s40001-025-02679-0] [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: 04/14/2025] [Accepted: 05/11/2025] [Indexed: 05/22/2025] Open
Abstract
Sepsis-induced lung injury is a serious complication that contributes to the high morbidity and mortality rates in septic patients. This study aims to identify genes associated with sepsis-induced lung injury and evaluate the role of cathepsin B (CTSB) in this process. Here, by analyzing three data sets of sepsis-induced lung injury in mouse models, we identified 23 common differentially expressed genes and performed enrichment analyses. Further experiments demonstrated that CTSB expression was significantly upregulated in the sepsis mouse model, and pre-treatment with the CTSB inhibitor CA-074 markedly improved the survival rate of the mice from 21.05 to 78.95%. In addition, the CTSB inhibitor reduced the systemic inflammatory response in septic mice by decreasing plasma levels of nitric oxide (NO) and the inflammatory cytokines TNF-α and IL-1β.Histological analysis showed that the CTSB inhibitor effectively suppressed CLP-induced lung tissue alterations and neutrophil infiltration, and significantly reduced the expression of inducible nitric oxide synthase (iNOS). Analysis of cell death indicated that the CTSB inhibitor decreased cell death in the lung tissue of CLP mice, particularly by inhibiting the upregulation of gasdermin D-N (GSDMD-N), which is associated with pyroptosis. Furthermore, in vitro experiments revealed that overexpression of CTSB enhanced cell death and promoted pyroptosis in lung epithelial cells. These results indicate that CTSB plays a crucial role in sepsis-induced lung injury, potentially exacerbating the inflammatory response by promoting pyroptosis. Therefore, CTSB may be a potential therapeutic target for sepsis-induced lung injury.
Collapse
Affiliation(s)
- Xiaobo Zhang
- Department of Emergency, Hebei Medical University Third Hospital, Shijiazhuang, 050000, Hebei, People's Republic of China
| | - Zhuojun Deng
- Department of Emergency, Hebei Medical University Third Hospital, Shijiazhuang, 050000, Hebei, People's Republic of China.
| | - Xinyu Zhang
- Department of Emergency, Hebei Medical University Third Hospital, Shijiazhuang, 050000, Hebei, People's Republic of China
| | - Qian Xu
- Department of Emergency, Hebei Medical University Third Hospital, Shijiazhuang, 050000, Hebei, People's Republic of China
| | - Li Liu
- Department of Emergency, Hebei Medical University Third Hospital, Shijiazhuang, 050000, Hebei, People's Republic of China
| | - Dong Yang
- Department of Emergency, Hebei Medical University Third Hospital, Shijiazhuang, 050000, Hebei, People's Republic of China
| | - Zimeng Guo
- Department of Rehabilitation, Hebei Medical University Third Hospital , Shijiazhuang, Hebei, People's Republic of China
| |
Collapse
|
12
|
Gao J, Han L, Zhang Y, Zhang X, Fei X, Zhang M. Disulfiram alleviates epithelial barrier disruption in ozone-induced chronic obstructive pulmonary disease mouse models via inhibiting Gasdermin D-mediated pyroptosis. Int Immunopharmacol 2025; 159:114887. [PMID: 40403507 DOI: 10.1016/j.intimp.2025.114887] [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: 01/20/2025] [Revised: 05/02/2025] [Accepted: 05/13/2025] [Indexed: 05/24/2025]
Abstract
BACKGROUND Gasdermin D (GSDMD)-mediated pyroptosis drives inflammatory cytokine release in response to environmental triggers. Disulfiram (DSF), an FDA-approved anti-alcoholism drug, has been demonstrated to inhibit GSDMD pore formation. Although airway epithelial barrier dysfunction contributes to chronic obstructive pulmonary disease (COPD) progression, the role of GSDMD-dependent pyroptosis in ozone-induced pathogenesis, and the potential of DSF to inhibit this process, remain unexplored. METHODS We analyzed the expression levels of pyroptosis-related molecules in airway epithelial cells from COPD patients' samples obtained from the Gene Expression Omnibus (GEO) database and evaluated the potential therapeutic effects of DSF in a mouse model of COPD induced by chronic ozone exposure. RESULTS GSDMD was significantly upregulated in the airway epithelial cells of COPD patients. Chronic ozone exposure in mice elevated the cleaved form of GSDMD and reduced the expression of epithelial junctional proteins. DSF treatment effectively inhibited GSDMD-mediated pyroptosis and attenuated epithelial barrier disruption, leading to significant improvements in airway inflammation and lung function in both large and small airways. Furthermore, Gsdmd expression was negatively correlated with the tight junction protein Occludin and pulmonary function indices, including the ratio of FEV25 to FVC and MMEF. CONCLUSION Collectively, these findings revealed the role of GSDMD-mediated pyroptosis in epithelial barrier disruption of COPD and the potential application of DSF in the treatment of COPD.
Collapse
Affiliation(s)
- Jianwei Gao
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Lei Han
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Yingying Zhang
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Xue Zhang
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Xia Fei
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.
| | - Min Zhang
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China.
| |
Collapse
|
13
|
Xu Z, Tang C, Song X, Liu Z, Zhou J, Shi Q, Yu C, Xu C. High uric acid exacerbates nonalcoholic steatohepatitis through NLRP3 inflammasome and Gasdermin D-mediated pyroptosis. J Biol Chem 2025:110249. [PMID: 40398602 DOI: 10.1016/j.jbc.2025.110249] [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: 01/24/2025] [Revised: 04/30/2025] [Accepted: 05/05/2025] [Indexed: 05/23/2025] Open
Abstract
Hyperuricemia is independently associated with an increased risk of nonalcoholic steatohepatitis (NASH), but the underlying mechanisms responsible for this association remain unclear. We first analyzed the association between intrahepatic UA levels and gasdermin D (GSDMD)-mediated pyroptosis in vivo and in vitro. We subsequently generated hepatic-specific glucose transporter 9 (GLUT9)-knockout mice and GSDMD knockout (GSDMD-/-) mice to explore the role of intrahepatic UA in GSDMD-induced pyroptosis in NASH. We found that high intrahepatic UA levels were positively related to GSDMD-mediated pyroptosis in NASH mice. The inhibition of hepatic UA production by allopurinol alleviated hepatic inflammation and GSDMD-mediated pyroptosis in NASH mice. Hepatic-specific knockout of Glut9 significantly decreased intrahepatic UA levels, attenuated NOD-like receptor family pyrin domain containing 3 (NLRP3)-Caspase-1-GSDMD-mediated pyroptosis in hepatocytes, and ameliorated hepatic inflammation and fibrosis in different mouse models of NASH. Further experiments revealed that inhibiting the NLRP3/Caspase-1/GSDMD pathway obviously blocked UA-induced pyroptosis and inflammation in hepatocytes. Additionally, GSDMD deficiency markedly reversed hepatic inflammation and fibrosis in NASH mice. In conclusion, our results showed that high UA could induce NLRP3-Caspase1-GSDMD-mediated pyroptosis, thereby aggravating NASH in mice.
Collapse
Affiliation(s)
- Zixin Xu
- Department of Gastroenterology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Chenxi Tang
- Department of Gastroenterology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Xin Song
- Department of Gastroenterology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Zhening Liu
- Department of Gastroenterology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jiaming Zhou
- Department of Gastroenterology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Qiaojuan Shi
- Zhejiang Provincial Key Laboratory of Laboratory Animals and Safety Research, Hangzhou Medical College, Hangzhou, 310063, China.
| | - Chaohui Yu
- Department of Gastroenterology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| | - Chengfu Xu
- Department of Gastroenterology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| |
Collapse
|
14
|
Xiao Z, Xie J, Zhao X, Chen X, Lu Y, Xu Y, Wu M, An L, Li Q. Role of Pyroptosis in inflammatory bowel disease. Int Immunopharmacol 2025; 155:114619. [PMID: 40209313 DOI: 10.1016/j.intimp.2025.114619] [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: 01/30/2025] [Revised: 03/21/2025] [Accepted: 04/03/2025] [Indexed: 04/12/2025]
Abstract
Inflammatory bowel disease (IBD) is a serious chronic condition marked by persistent and recurrent intestinal ulcers. Although the exact cause of IBD remains unclear, it is generally accepted that a complex interaction among dietary factors, gut microbiota, and immune responses in genetically predisposed individuals contributes to its development. Pyroptosis, an inflammatory form of programmed cell death activated by inflammasomes, is marked by the rupture of cell membranes and the subsequent release of inflammatory mediators. Emerging evidence indicates that pyroptosis plays a crucial role in the pathogenesis of IBD. Moderate pyroptosis activation can enhance intestinal immune defenses, while excessive inflammasome activation can trigger an inflammatory cascade, resulting in increased damage to intestinal tissues. This article reviews the molecular mechanisms underlying pyroptosis and highlights its role in the onset and progression of IBD. Furthermore, We explore recent advancements in IBD treatment, focusing on small molecule compounds that specifically target and inhibit pyroptosis.
Collapse
Affiliation(s)
- Zhiyi Xiao
- The Clinical Medical College, Guizhou Medical University, Guiyang 550004, China
| | - Jiling Xie
- The Clinical Medical College, Guizhou Medical University, Guiyang 550004, China
| | - Xun Zhao
- Department of Gastroenterology, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, China
| | - Xiangjun Chen
- The Clinical Medical College, Guizhou Medical University, Guiyang 550004, China
| | - Yihong Lu
- The Clinical Medical College, Guizhou Medical University, Guiyang 550004, China
| | - Yuanzhao Xu
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, China
| | - Manqing Wu
- Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, China
| | - Lingyue An
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, China.
| | - Qing Li
- Department of Gastroenterology and Surgery, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, China.
| |
Collapse
|
15
|
Zhang H, Song L, Zhou L, Li X, Xuan M, Liu C, Zhao H. α -Lipoic acid alleviates Parkinson's disease by suppressing S100A9-mediated pyroptosis. Int Immunopharmacol 2025; 155:114539. [PMID: 40233449 DOI: 10.1016/j.intimp.2025.114539] [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: 01/08/2025] [Revised: 03/17/2025] [Accepted: 03/20/2025] [Indexed: 04/17/2025]
Abstract
Parkinson's disease (PD) is a neurodegenerative disease, and inflammation is a key factor in the progression of PD. S100A9 mediates pyroptosis and implicates in various diseases including PD. Pyroptosis, an emerging form of programmed cell death, usually causes cell rupture and death via an inflammatory response. α-Lipoic acid (α-ALA), a cellular coenzyme, participates in anti-inflammatory and antioxidant processes. Although its role in PD has been confirmed, but the exact mechanism of its anti-inflammatory effect remains unclear. In our research, we examined the potential mechanisms of pyroptosis mediated by S100A9 in PD and the neuroprotective effects of α-ALA. We used 6-hydroxydopamine (6-OHDA) to induce SH-SY5Y cells in vitro and in C57BL/6 mice in vivo. The cell viability of SH-SY5Y cells confirmed the neuroprotective effect of α-ALA. Proteomics analysis indicated that S100A9 was involved in 6-OHDA-mediated neuronal injury, while α-ALA could inhibit. We found that α-ALA ameliorated PD symptoms induced by 6-OHDA and decreased the levels of NLRP3 inflammasome, Gasdermin D, and IL-1β, which are major hallmarks of pyroptosis. Furthermore, our research demonstrated that α-ALA mitigated cell injury by suppressing NLRP3-dependent pyroptosis mediated by S100A9. In brief, pyroptosis is pivotal in PD, while α-ALA protects dopaminergic neurons by suppressing pyroptosis mediated through the NLRP3 inflammasome, directly reducing S100A9, and subsequently inhibiting the NLRP3/Gasdermin D signaling pathways. Our results collectively suggest that suppressing S100A9-mediated pyroptosis and administering α-ALA may represent a novel approach in treating of PD.
Collapse
Affiliation(s)
- Hongxu Zhang
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Ling Song
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Lin Zhou
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Xiaoyuan Li
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Mingwen Xuan
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Chang Liu
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, China.
| | - Hong Zhao
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, China.
| |
Collapse
|
16
|
Zhang S, Zhu X, Yu W, Yu Y, Qian L, Chen Z, Peng Z, Gao L, Chen L, Chen J. Self-Assembly of Ru3-Aptamer Nanoparticles Triggers Pyroptosis through Photoredox Catalysis of NADH and Lysosomal Disruption. J Med Chem 2025. [PMID: 40372006 DOI: 10.1021/acs.jmedchem.5c00022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
Abstract
Photodynamic therapy (PDT) can induce tumor cell death. Ru3, a metal-based photosensitizer, features a high positive charge, a long triplet excited-state lifetime, and an excellent PDT activity. The aptamer AS1411, known for its ability to selectively bind to nucleolin (which is overexpressed in tumor cells), self-assembled with Ru3 into nanoparticles termed Ru3ApNPs. These nanoparticles specifically target SiHa tumor cells. Upon light irradiation, Ru3ApNPs increase intracellular ROS levels, catalyze NADH redox reactions, and induce lysosomal disruption, ultimately triggering pyroptosis in tumor cells. Notably, Ru3ApNPs demonstrate excellent tumor penetration in 3D multicellular spheroids (MCSs) of SiHa cells and effectively inhibit their growth under light exposure. Ru3ApNPs exhibit a mechanism of action distinct from that of traditional PDT. Furthermore, under light irradiation, Ru3ApNPs can effectively inhibit the growth of distant tumors and induce systemic immune responses in mice. Our data suggest that Ru3ApNPs can be developed as promising targeted therapeutic agents in the future.
Collapse
Affiliation(s)
- Shenting Zhang
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong 524023, P. R. China
| | - Xufeng Zhu
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong 524023, P. R. China
| | - Wenzhu Yu
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong 524023, P. R. China
| | - Yunjiang Yu
- School of Chemistry and Chemical Engineering, Lingnan Normal University, Zhanjiang 524048, P. R. China
| | - Li Qian
- Youjiang Medical College for Nationalities, Baise 533000, P. R. China
| | - Zhikai Chen
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong 524023, P. R. China
| | - Zitong Peng
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong 524023, P. R. China
| | - Lijun Gao
- School of Chemistry and Chemical Engineering, Lingnan Normal University, Zhanjiang 524048, P. R. China
| | - Lanmei Chen
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong 524023, P. R. China
| | - Jincan Chen
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, Guangdong 524023, P. R. China
| |
Collapse
|
17
|
Hu J, Niu J, Jiang S, Wu Y. Qilian Jiechang Ning Alleviates TNBS-Induced Ulcerative Colitis in Mice and Segatella copri Outer Membrane Vesicle-Triggered Inflammation in Colon Epithelial Cells via the Caspase-1/11-GSDMD Pathways. J Innate Immun 2025; 17:262-276. [PMID: 40367931 PMCID: PMC12077867 DOI: 10.1159/000545394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 03/18/2025] [Indexed: 05/16/2025] Open
Abstract
INTRODUCTION Qilian Jiechang Ning (QJN), a traditional Chinese herbal formula, has demonstrated potential therapeutic effects in the treatment of ulcerative colitis (UC). This study aims to investigate the mechanism of QJN in the outer membrane vesicles (OMVs) of Segatella copri (S. copri)-induced colon epithelial cells and UC mice. METHODS Transmission electron microscopy (TEM) and nanoparticle tracking analysis (NTA) were utilized to assess the morphology and size of OMVs. Inflammation markers and tight junction protein levels in HCoEpiCs induced by OMVs were monitored using ELISA and western blot. QJN was administered to intervene in HCoEpiCs treated with S. copri OMVs. Additionally, trinitrobenzene sulfonic acid (TNBS)-induced mouse models were conducted to evaluate the therapeutic effects of QJN on UC. RESULTS S. copri OMVs treated with QJN demonstrated a significant reduction in particle size, protein concentration, and LPS content. In HCoEpiCs, QJN effectively decreased the expression of inflammation-inducing cytokines (IL-1β, IL-18, IL-6, TNF-α) and proinflammatory proteins (GSDMD-N, NLRP3, ASC, cleaved Caspase-1, cleaved Caspase-4) triggered by S. copri OMVs, while enhancing the expression of tight junction proteins (ZO-1 and Occludin). In the UC mouse models, QJN significantly reduced the Disease Activity Index (DAI), improved colon length, lowered LPS levels, ameliorated colonic tissue damage, and inhibited Caspase-1- and Caspase-11-dependent inflammatory responses. CONCLUSION QJN can alleviate S. copri-OMV-induced inflammatory response in colonic epithelial cells and reduce symptoms of UC in mouse models by modulating the Caspase-1 and Caspase-11 pathways.
Collapse
Affiliation(s)
- Jinyang Hu
- Department of Spleen, Gastroenterology and Hepatology, Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine, Changsha, PR China
| | - Junjie Niu
- Department of Radiation Oncology, Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine, Changsha, PR China
| | - Shisheng Jiang
- Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine, Changsha, PR China
| | - Yuhua Wu
- Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine, Changsha, PR China
| |
Collapse
|
18
|
Li J, Li Y, Chen Z, Yang L, Zhao L, Li J. PINK1 activation by MTK458 ameliorates neurological impairments and pyroptosis after intracerebral hemorrhage in mice. Brain Res 2025; 1861:149700. [PMID: 40368226 DOI: 10.1016/j.brainres.2025.149700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Revised: 04/26/2025] [Accepted: 05/09/2025] [Indexed: 05/16/2025]
Abstract
Intracerebral hemorrhage (ICH) is often linked to severe neurological impairments, including cognitive deficits and anxiety-like behaviors. This study aimed to evaluate the therapeutic potential of PTEN-induced kinase 1 (PINK1), which is activated during ICH, as a target for mitigating these effects. C57/BL6 wild-type mice underwent ICH induction through an intrastriatal injection of autologous blood. The PINK1 activator, MTK458, was administered daily doses of 10-50 mg/kg starting one week before ICH induction and continuing for three days post-surgery. The modified neurological severity score (mNSS) was used to assess neurological deficits, while brain edema was measured through brain water content. The open field test and Y-maze test were used to evaluate anxiety-like behavior, and cognitive function respectively. The effects of ICH on cortical cell pyroptosis, Parkin/PINK1-mediated mitophagy, and the activation of the NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasome were analyzed via Western blotting, ELISA, and qRT-PCR. MTK458 effectively reduced brain water content in the basal ganglia, ipsilateral cortex, and cerebellum, with improvements in mNSS extending to 14 days post-injury. Additionally, MTK458 alleviated both neurological deficits and anxiety-like behavior in ICH mouse models. It also reversed ICH-induced cortical cell pyroptosis by promoting Parkin/PINK1-mediated mitophagy and inhibiting NLRP3 inflammasome activation, as well as the expression of IL-1β and IL-18. These results suggest that MTK458 effectively reduces neurological impairments, brain edema, and anxiety-related behaviors in mice following ICH, highlighting PINK1 activation as a promising therapeutic strategy for ICH-induced neurological deficits.
Collapse
Affiliation(s)
- Jianliang Li
- Department of Neurosurgery, the Second Hospital of Hebei Medical University, No.215 Hepingxi Road, Shijiazhuang 050000 Hebei, China
| | - Yincheng Li
- Department of Emergency Medicine, the First Hospital of Hebei Medical University, No.89 Donggang Road, Shijiazhuang 050000 Hebei, China
| | - Zhe Chen
- Department of Cardiovascular Medicine, the Third Hospital of Hebei Medical University, No.139 Ziqiang Road, Shijiazhuang 050000 Hebei, China
| | - Liang Yang
- Department of Neurosurgery, the Second Hospital of Hebei Medical University, No.215 Hepingxi Road, Shijiazhuang 050000 Hebei, China
| | - Lin Zhao
- Department of Neurosurgery, the Second Hospital of Hebei Medical University, No.215 Hepingxi Road, Shijiazhuang 050000 Hebei, China
| | - Jingchen Li
- Department of Neurosurgery, the Second Hospital of Hebei Medical University, No.215 Hepingxi Road, Shijiazhuang 050000 Hebei, China.
| |
Collapse
|
19
|
Chen J, Xu M, Wu F, Wu N, Li J, Xie Y, Wang R, Xi N, Zhu Y, Xu X, Liu Y. CRKL silencing inhibits melanoma growth and enhances its chemotherapy sensitivity through the PI3K/AKT and NLRP3/GSDMD pathways. Biochem Pharmacol 2025; 235:116840. [PMID: 40024349 DOI: 10.1016/j.bcp.2025.116840] [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: 11/11/2024] [Revised: 01/27/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
Great advances have been made in malignant melanoma treatments, whereas drug resistance still limits many drug applications. CRKL has been reported to be overexpressed in various tumors and showed poor prognosis. However, its specific function and mechanism in melanoma remain unclear. In the present study, we investigated the expression of CRKL and its clinical association by bioinformatics and clinical analysis, and then performed a series of in vitro and in vivo experiments to demonstrate its function and mechanism. Results showed that CRKL increased during melanoma progression and was strongly associated with poor prognosis. CRKL silencing effectively inhibited melanoma cell growth and invasion via ERK/MMP9 and PI3K/AKT signaling pathways both in vitro and in vivo. Moreover, CRKL silencing induced pyroptosis in melanoma cells by upregulating the levels of pyroptosis-associated proteins, such as NLRP3, cleaved Caspase-1, and GSDMD-N. Importantly, our study demonstrated that interfering with CRKL expression enhanced the chemotherapy sensitivity of melanoma cells to cisplatin by regulating PI3K/AKT and NLRP3/GSDMD signaling pathways. In conclusion, our study uncovers a novel molecular mechanism by which CRKL functions in melanoma and highlights potential therapeutic strategies for improving chemotherapy sensitivity in melanoma patients.
Collapse
Affiliation(s)
- Jiashe Chen
- Department of Pathology, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China
| | - Mingyuan Xu
- Department of Pathology, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China
| | - Fei Wu
- Department of Pathology, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China
| | - Nanhui Wu
- Department of Pathology, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China
| | - Jie Li
- Department of Pathology, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China
| | - Yongyi Xie
- Department of Pathology, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China
| | - Ruoqi Wang
- Department of Pathology, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China
| | - Ningyuan Xi
- Department of Pathology, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China
| | - Yueyi Zhu
- Department of Pathology, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China
| | - Xiaoxiang Xu
- Department of Pathology, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China.
| | - Yeqiang Liu
- Department of Pathology, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China.
| |
Collapse
|
20
|
Lu XY, Zhu LY, Zhu H, Huang SJ, Yang YS, Jiang CR, Ye RR. Cyclometalated iridium(III)-lonidamine conjugates: Mitochondrial targeting and pyroptosis induction. J Inorg Biochem 2025; 266:112852. [PMID: 39938148 DOI: 10.1016/j.jinorgbio.2025.112852] [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: 12/27/2024] [Revised: 01/27/2025] [Accepted: 02/05/2025] [Indexed: 02/14/2025]
Abstract
A series of cyclometalated Ir(III)-lonidamine (LND) complexes (Ir-LND-1-6) with the formula [Ir(C^N)2bpy(4-CH3-4'-CH2OLND)](PF6) (Ir-LND-1-3) and [Ir(C^N)2bpy(4-CH2OLND-4'-CH2OLND)](PF6) (Ir-LND-4-6) (C^N = 2-phenylpyridine (ppy, in Ir-LND-1 and Ir-LND-4), 2-(2-thienyl) pyridine (thpy, in Ir-LND-2 and Ir-LND-5) and 2-(2,4-difluorophenyl) pyridine (dfppy, in Ir-LND-3 and Ir-LND-6)), were designed and synthesized. 3-(4,5-dimethylthiazol-2-yl)-2,5-biphenyltetrazolium bromide (MTT) assay data showed that the cytotoxicity of Ir-LND-1-3 carry one LND moiety was superior to that of Ir-LND-4-6 with two LND moieties. Therefore, we selected Ir-LND-1-3 as model compounds to investigate the anti-tumor mechanism of the Ir(III)-LND system. The results showed that Ir-LND-1-3 could inhibit cancer cell migration and colony formation. In addition, Ir-LND-1-3 could penetrate into HeLa cells and localized to mitochondria, further disrupting mitochondrial membrane potential (MMP), increasing intracellular reactive oxygen species (ROS), and reducing intracellular adenosine triphosphate (ATP). Further exploration of anti-tumor mechanisms showed that pyroptosis was the main mode of Ir-LND-1-3 induced cell death, manifested as membrane perforation and swelling, activation of caspase-3 and cleavage of Gasdermin E (GSDME), as well as release of lactic dehydrogenase (LDH) and ATP. The pyroptosis induced by Ir-LND-1-3 also initiated immunogenic cell death (ICD) by triggering the release of calreticulin (CRT) and high mobility group protein b1 (HMGB1) on the cell surface.
Collapse
Affiliation(s)
- Xing-Yun Lu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, PR China
| | - Lin-Yuan Zhu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, PR China
| | - Hou Zhu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, PR China
| | - Shao-Jun Huang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, PR China.
| | - Yong-Sheng Yang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, PR China
| | - Chun-Rong Jiang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, PR China
| | - Rui-Rong Ye
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, PR China.
| |
Collapse
|
21
|
Ju X, Zhang H, Wang J, Wang J, Wang N, Geng J, Guo L, Wang Q. LXR agonists induces GSDME-mediated pyroptosis in tumors through alters the integrity of the MOM to activates Caspase-4/APAF-1 pyroptosome. Int J Biol Macromol 2025; 310:142568. [PMID: 40154703 DOI: 10.1016/j.ijbiomac.2025.142568] [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: 10/26/2024] [Revised: 03/24/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
Liver X receptors (LXRs) play a key role in cholesterol transport, glucose metabolism and the tumorigenesis. LXR ligands can inhibit tumor growth through several mechanisms, such as suppressing tumor cell proliferation and migration, and inducing tumor cell death. Among them, induction of tumor cell death is one of the main mechanisms by which LXR ligands inhibit tumor growth; but how exactly LXR ligands cause cell death is still unclear. In this study, we found that LXR agonists can induce nonclassical pyroptosis in various cancer cells. Further study we found that Caspase-3-mediated GSDME cleavage is involved in LXR agonist-induced pyroptosis. Mechanically, LXR agonists firstly induce ER stress in tumor cells, then the ER stress induced by LXR agonists alters the integrity of the mitochondrial outer membrane (MOM) through the NOXA and BAX/BAK. Subsequently, mitochondrial permeability transition activates Caspase-4/APAF-1 pyroptosome to activate GSDME-dependent pyroptosis. Finally, we also found that LXR agonists induced GSDME-dependent pyroptosis in mouse cells. Our results demonstrated that LXR agonists can induce nonclassical GSDME-dependent pyroptosis in cancer cells by inducing ER stress, which alters the integrity of MOM to activate Caspase-4/APAF-1 pyroptosome.
Collapse
Affiliation(s)
- Xiaoli Ju
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Heng Zhang
- Department of General Surgery, Nanjing Lishui District People's Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing, China
| | - Jin Wang
- Institute of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Jiayou Wang
- Institute of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Ning Wang
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Jingyao Geng
- School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Lanfang Guo
- Department of Clinical Laboratory Medicine, The Fourth People's Hospital of Jiangsu University, Zhenjiang, Jiangsu 212013, China.
| | - Qiang Wang
- Institute of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China.
| |
Collapse
|
22
|
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
|
23
|
Zhan Q, Kuang Y, Chen X, Yang Y, Jiang L, Chen J, Li L, Wang J, Zhu S, Huang H, Wang L, Zhu P, Liu R. Photo-generating Type-I ROS and aryl radicals by mitochondrial-targeting oxime-ester photogenerator for pyroptosis-mediated anti-hypoxia photoimmunotherapy. Bioact Mater 2025; 47:327-342. [PMID: 40026820 PMCID: PMC11870024 DOI: 10.1016/j.bioactmat.2025.01.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 01/23/2025] [Accepted: 01/23/2025] [Indexed: 03/05/2025] Open
Abstract
Pyroptosis is an inflammatory form of programmed cell death with great potential in cancer immunotherapies. Photodynamic therapy (PDT) represents a promising treatment modality to trigger pyroptosis. However, the hypoxic microenvironment inside the tumors often induces limited therapeutic efficacy. Herein, in this work, the first type of mitochondrial-targeting oxime-ester photogenerator (T-Oximer) was constructed to boost type-I ROS/aryl free radicals which could induce DNA damage by DNA cleaving and facilitate high-efficiency pyroptosis-mediated photoimmunotherapy. Detailed mechanism investigations revealed that T-Oximer could produce aryl free radicals via photolysis reaction and generate type-I ROS (O2 •- and •OH) based on the type-I electron transfer process. Meanwhile, T-Oximer could accumulate in the mitochondria, boost mitochondrial radicals, and damage mitochondria in hypoxic tumor cells. Of peculiar interest, T-Oixmer could bind with DNA and cleave DNA to induce DNA damage. Combined mitochondrial damage with DNA cleavage, T-Oximer can initiate pyroptosis, activate the ICD effect, and trigger robust systemic antitumor immunity for efficient tumor regression and metastasis suppression. Our finding provides a new strategy for constructing oxygen-independent photogenerator for high-efficiency pyroptosis-mediated anti-hypoxia photoimmunotherapy.
Collapse
Affiliation(s)
- Qiyu Zhan
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, China
| | - Yulin Kuang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, Guangdong, 510100, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510006, China
| | - Xuyuan Chen
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, Guangdong, 510100, China
- Comprehensive Medical Treatment Ward, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yanzhen Yang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, Guangdong, 510100, China
| | - Linhui Jiang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, Guangdong, 510100, China
| | - Jian Chen
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, China
| | - Lie Li
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, China
| | - Junwei Wang
- Department of Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou Avenue North No. 1838, Baiyun District, Guangzhou, 510515, China
| | - Shuoji Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, Guangdong, 510100, China
| | - Huanlei Huang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial People's Hospital Ganzhou Hospital, Ganzhou, 341000, China
| | - Lei Wang
- College of Materials and Chemical Engineering, Key Laboratory of Inorganic Nonmetallic Crystalline and Energy Conversion Materials, China Three Gorges University, Yichang, 443002, China
| | - Ping Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, Guangdong, 510100, China
- Guangdong Provincial People's Hospital Ganzhou Hospital, Ganzhou, 341000, China
| | - Ruiyuan Liu
- Biomaterials Research Center, School of Biomedical Engineering, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
24
|
Liang JL, Cao Y, Lv K, Xiao B, Sun J. Amplifying Ca 2+ overload by engineered biomaterials for synergistic cancer therapy. Biomaterials 2025; 316:123027. [PMID: 39700532 DOI: 10.1016/j.biomaterials.2024.123027] [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: 10/07/2024] [Revised: 11/28/2024] [Accepted: 12/13/2024] [Indexed: 12/21/2024]
Abstract
Ca2+ overload is one of the most widely causes of inducing apoptosis, pyroptosis, immunogenic cell death, autophagy, paraptosis, necroptosis, and calcification of tumor cells, and has become the most valuable therapeutic strategy in the field of cancer treatment. Nevertheless, several challenges remain in translating Ca2+ overload-mediated therapeutic strategies into clinical applications, such as the precise control of Ca2+ dynamics, specificity of Ca2+ homeostasis dysregulation, as well as comprehensive mechanisms of Ca2+ regulation. Given this, we comprehensively reviewed the Ca2+-driven intracellular signaling pathways and the application of Ca2+-based biomaterials (such as CaCO3-, CaP-, CaO2-, CaSi-, CaF2-, and CaH2-) in mediating cancer diagnosis, treatment, and immunotherapy. Meanwhile, the latest researches on Ca2+ overload-mediated therapeutic strategies, as well as those combined with multiple-model therapies in mediating cancer immunotherapy are further highlighted. More importantly, the critical challenges and the future prospects of the Ca2+ overload-mediated therapeutic strategies are also discussed. By consolidating recent findings and identifying future research directions, this review aimed to advance the field of oncology therapy and contribute to the development of more effective and targeted treatment modalities.
Collapse
Affiliation(s)
- Jun-Long Liang
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China.
| | - Yangyang Cao
- Hangzhou Ultra-theranostics Biopharmaceuticals Technology Co., Ltd., Hangzhou, 311231, China
| | - Kaiwei Lv
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| | - Bing Xiao
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China.
| | - Jihong Sun
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China; Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, China.
| |
Collapse
|
25
|
Li Q, Wang Z, Li F, Liu S, Ding Y, Yan J, Feng X, Li M. AIM2 exacerbates hypoxic-ischemic brain damage in neonatal rats via promoting neuronal pyroptosis. Brain Res Bull 2025; 224:111305. [PMID: 40101806 DOI: 10.1016/j.brainresbull.2025.111305] [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: 12/23/2024] [Revised: 03/03/2025] [Accepted: 03/12/2025] [Indexed: 03/20/2025]
Abstract
BACKGROUND Pyroptosis has been reported to play a pathogenic role in neonatal hypoxic-ischemic brain damage (HIBD). Absence in melanoma 2 (AIM2) is an inflammasome involved in pyroptosis. OBJECTIVE This study aimed to investigate the role of AIM2 in hypoxic-ischemia (HI)-induced pyroptosis and brain damage in a neonatal rat HIBD model. METHODS In vivo, we injected a lentivirus that overexpressed or knocked down AIM2 into the lateral ventricle of rats within 24 h after birth and prepared a 7-day Sprague Dawley (SD) rat HIBD model. In vitro, we transfected lentiviruses overexpressing or knocking down AIM2 into cultured primary neurons and established an oxygen/glucose deprivation/reoxygenation (OGD/R) model. 2,3,5-triphenyltetrazolium chloride (TTC) staining was used to determine infarct size. Hematoxylin and eosin and Nissl staining were used to evaluate morphological changes in the damaged brain. Cell Counting Kit-8 (CCK-8) and lactate dehydrogenase (LDH) assays were used to determine cell viability and toxicity. Pyroptosis was observed using transmission electron microscopy. RESULTS AIM2 expression significantly increased in the HI-induced cortex of neonatal rats. Lentivirus-mediated overexpression of AIM2 significantly aggravates HI-induced brain injury and OGD/R-induced neuronal injury in vivo and in vitro. The lentivirus-mediated AIM2 knockdown significantly reversed these adverse effects. In addition, AIM2 overexpression increased HI-induced pyroptosis in neonatal rats in vivo and in vitro, whereas AIM2 knockdown suppressed HI-induced pyroptosis via the AIM2/Caspase-1/GSDMD pathway. CONCLUSION These findings show that the upregulation of AIM2 activates pyroptosis and plays a pathogenic role in neonatal HIBD.
Collapse
Affiliation(s)
- Qianqian Li
- Department of Neonatology, Xuzhou Maternity and Child Health Care Hospital, Xuzhou, Jiangsu Province, China; Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Zengqin Wang
- Department of Neonatology, Xuzhou Maternity and Child Health Care Hospital, Xuzhou, Jiangsu Province, China
| | - Fengli Li
- Department of Intensive Care Unit, Zibo Central Hospital, Zibo, Shandong Province, China
| | - Songlin Liu
- Department of Neonatology, Xuzhou Maternity and Child Health Care Hospital, Xuzhou, Jiangsu Province, China
| | - Yuhong Ding
- Department of Neonatology, Xuzhou Maternity and Child Health Care Hospital, Xuzhou, Jiangsu Province, China
| | - Junmei Yan
- Department of Neonatology, Xuzhou Maternity and Child Health Care Hospital, Xuzhou, Jiangsu Province, China.
| | - Xing Feng
- Soochow Key Laboratory of Prevention and Treatment of Child Brain Injury, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, China.
| | - Mei Li
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, China.
| |
Collapse
|
26
|
Jin H, Wu P, Lv C, Zhang S, Zhang Y, Li C, Gao R, Shan G, Bi H, Chang H, Liu X, Zeng Y. Mannose inhibits PKM2 lactylation to induce pyroptosis in bladder cancer and activate antitumor immune responses. Commun Biol 2025; 8:689. [PMID: 40312519 PMCID: PMC12045973 DOI: 10.1038/s42003-025-08130-8] [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: 12/02/2024] [Accepted: 04/25/2025] [Indexed: 05/03/2025] Open
Abstract
Bladder cancer therapy remains challenging due to poor efficacy and frequent recurrence. Mannose, a naturally occurring monosaccharide, has demonstrated antitumor effects in various cancers, yet its mechanism of action in bladder cancer is unclear. This study explored the inhibitory effects of mannose on bladder cancer. We found mannose significantly inhibited the growth of bladder cancer cells, xenografts, and organoids. Mannose directly binds to PKM2, inhibiting its enzymatic activity and reducing lactate production. This reduction in lactate led to decreased PKM2 lactylation and increased acetylation, causing PKM2 to translocate to the nucleus. Nuclear PKM2 activated the NF-κB pathway, inducing NLRP1/Caspase-1/GSDMD/IL-1β-dependent pyroptosis. Additionally, mannose promoted antitumor immune responses by inducing pyroptosis and enhancing the efficacy of immune checkpoint inhibitors. These findings highlight the use of mannose as a potent antitumor agent and a promising therapeutic strategy for bladder cancer.
Collapse
Affiliation(s)
- Haoyi Jin
- Department of Thoracic Surgery and Oncology, the First Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
- Department of Thoracic Surgery and Oncology, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning Province, China
| | - Pingeng Wu
- Department of Urology, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning Province, China
| | - Chengcheng Lv
- Department of Urology, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning Province, China
| | - Shouyi Zhang
- Department of Urology, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning Province, China
| | - Yunchao Zhang
- Department of Urology, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning Province, China
| | - Changqi Li
- Department of Urology, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning Province, China
| | - Ruxu Gao
- Department of Urology, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning Province, China
| | - Guangyi Shan
- Department of Urology, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning Province, China
| | - Huan Bi
- Department of Urology, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning Province, China
| | - Hong Chang
- Department of Anesthesiology, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Xi Liu
- Department of Urology, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning Province, China.
| | - Yu Zeng
- Department of Urology, Cancer Hospital of China Medical University, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning Province, China.
| |
Collapse
|
27
|
Fang F, Tang J, Geng J, Fang C, Zhang B. N-acetylserotonin derivative ameliorates hypoxic-ischemic brain damage by promoting PINK1/Parkin-dependent mitophagy to inhibit NLRP3 inflammasome-induced pyroptosis. Int Immunopharmacol 2025; 153:114469. [PMID: 40106901 DOI: 10.1016/j.intimp.2025.114469] [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: 10/13/2024] [Revised: 01/12/2025] [Accepted: 03/09/2025] [Indexed: 03/22/2025]
Abstract
Neonatal hypoxic-ischemic brain damage is the main cause of hypoxic-ischemic encephalopathy and cerebral palsy, whose clinical treatment is still limited to therapeutic hypothermia with limited efficacy. N-[2-(5-hydroxy-1H-indol-3-yl) ethyl]-2-oxopiperidine-3-carboxamide (HIOC), a derivative of N-acetylserotonin, has shown neuroprotective properties. This study was conducted to evaluate the neuroprotective and molecular mechanisms of HIOC. We established an in vitro model using Oxygen-glucose deprivation/reoxygenation (OGD/R) in HT22 cells, alongside an in vivo model via the modified Rice-Vannucci method. The results showed that HIOC reduced OGD/R-induced HT22 cell pyroptosis and inhibited NOD-like receptor pyrin domain- containing protein 3 (NLRP3) inflammasome activation. With the addition of the mitophagy inhibitor 3-MA, we demonstrated that HIOC promoted PTEN-induced putative kinase 1 (PINK1)/Parkin-mediated mitophagy to reduce HT22 cell pyroptosis. Mechanistically, HIOC stimulated mitophagy to remove damaged mitochondria. The clearance of injured mitochondria reduced reactive oxygen species generation, which consequently inhibited NLRP3 inflammasome expression. In vivo, HIOC remarkably lessened cerebral blood flow, infarct volume, neuronal injury by activating mitophagy. HIOC activated mitophagy to produce antipyroptosis effects. Together, our finding demonstrated that HIOC improves brain injury by promoting PINK1/Parkin-dependent mitophagy to inhibit NLRP3 inflammasome activation and pyroptosis, suggesting its potential for hypoxic-ischemic brain damage treatment.
Collapse
Affiliation(s)
- Fang Fang
- Departments of Neonatology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jiaxin Tang
- Departments of Neonatology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China; Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Jiaqing Geng
- Departments of Neonatology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Chengzhi Fang
- Departments of Neonatology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| | - Binghong Zhang
- Departments of Neonatology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
28
|
Wu W, Lan W, Jiao X, Wang K, Deng Y, Chen R, Zeng R, Li J. Pyroptosis in sepsis-associated acute kidney injury: mechanisms and therapeutic perspectives. Crit Care 2025; 29:168. [PMID: 40270016 PMCID: PMC12020238 DOI: 10.1186/s13054-025-05329-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 02/21/2025] [Indexed: 04/25/2025] Open
Abstract
Sepsis-associated acute kidney injury (S-AKI) is a severe complication characterized by high morbidity and mortality, driven by multi-organ dysfunction. Recent evidence suggests that pyroptosis, a form of programmed cell death distinct from apoptosis and necrosis, plays a critical role in the pathophysiology of S-AKI. This review examines the mechanisms of pyroptosis, focusing on inflammasome activation (e.g., NLRP3), caspase-mediated processes, and the role of Gasdermin D in renal tubular damage. We also discuss the contributions of inflammatory mediators, oxidative stress, and potential therapeutic strategies targeting pyroptosis, including inflammasome inhibitors, caspase inhibitors, and anti-inflammatory therapies. Lastly, we highlight the clinical implications and challenges in translating these findings into effective treatments, underscoring the need for personalized medicine approaches in managing S-AKI.
Collapse
Affiliation(s)
- Wenyu Wu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- Guangdong Clinical Research Academy of Chinese Medicine, Guangzhou, 510405, China
| | - Wanning Lan
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Xin Jiao
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, 510120, China
| | - Kai Wang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Yawen Deng
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, 510120, China
| | - Rui Chen
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Research On Emergency in TCM, Guangzhou, Guangdong, China.
| | - Ruifeng Zeng
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Research On Emergency in TCM, Guangzhou, Guangdong, China.
| | - Jun Li
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
- The First Clinical Medical School of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
| |
Collapse
|
29
|
Li Y, Han L, Hu H. Research progress on cuproptosis and copper related anti-tumor therapy. Discov Oncol 2025; 16:584. [PMID: 40257639 PMCID: PMC12011693 DOI: 10.1007/s12672-025-02335-3] [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: 04/06/2024] [Accepted: 04/08/2025] [Indexed: 04/22/2025] Open
Abstract
Copper is a trace element which is essential for biological organisms, and its homeostatic balance is important for living organisms to maintain the normal function. When the copper homeostasis is disordered, the cellular function and structure will be disrupted. Excess copper cause oxidative stress and DNA damage in cells, thereby inducing regulated cell death such as apoptosis and necroptosis. Excess copper in mitochondria can bind to lipoylated proteins in the tricarboxylic acid (TCA) cycle and cause them to aggregate, resulting in proteotoxic stress and eliciting a novel cell death modality: cuproptosis. Cancer cells have a greater demand for copper compared to normal tissue, and high levels of copper ions are closely associated with tumour proliferation and metastasis. The anti-tumor mechanisms of copper include the production of oxidative stress, inhibition of the ubiquitin-proteasome system, suppression of angiogenesis, and induction of copper-dependent cell death. Targeting copper is one of the current directions in oncology research, including the use of copper ion carriers to increase intracellular copper levels to induce oxidative stress and cuproptosis, as well as the use of copper ion chelators to reduce copper bioavailability. However, copper complexes have certain toxicity, so their biosafety needs to be improved. Emerging nanotechnology is expected to solve this problem by utilizing copper-based nanomaterials (Cu-based NMs) to deliver copper ions and a variety of drugs with different functions, thereby improving the anti-tumor efficacy and reducing the side effects. Therefore, a thorough understanding of copper metabolic processes and the mechanism of cuproptosis will greatly benefit anti-tumor therapy. This review summarizes the processes of copper metabolism and the mechanism of cuproptosis. In addition, we discuss the current anti-tumor paradigms related to copper, we also discuss current nanotherapeutic approaches to copper mortality and provide prospective insights into the future copper-mediated cancer therapy.
Collapse
Affiliation(s)
- Yichen Li
- School of Medicine, Southeast University, No. 87, Dingjiaqiao, Hunan Road, Gulou District, Nanjing, 210009, China
| | - Lifei Han
- Breast Disease Diagnosis and Treatment Center, Zhongda Hospital Affiliated to Southeast University, Nanjing, 210009, China
| | - Haolin Hu
- Breast Disease Diagnosis and Treatment Center, Zhongda Hospital Affiliated to Southeast University, Nanjing, 210009, China.
| |
Collapse
|
30
|
Song S, Wang J, Ouyang X, Huang R, Wang F, Xie J, Chen Q, Hu D. Therapeutic connections between pyroptosis and paclitaxel in anti-tumor effects: an updated review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04036-8. [PMID: 40257490 DOI: 10.1007/s00210-025-04036-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 03/06/2025] [Indexed: 04/22/2025]
Abstract
As a form of inflammation-associated cell death, pyroptosis has gained widespread attention in recent years. Accumulating evidence indicates that pyroptosis regulates tumor growth and is associated with autoimmune disorders and inflammatory response. Paclitaxel, a traditional Chinese medicine, usually induces death of cancer cells as a chemotherapeutic agent. Previous studies have revealed that paclitaxel can exert an anti-tumor effect through a variety of cell death mechanisms, of which pyroptosis plays a pivotal role in inhibiting tumor growth and enhancing anti-tumor immunity. In this review, we summarize the current advances in therapeutic connections between pyroptosis and paclitaxel in anti-tumor effects.
Collapse
Affiliation(s)
- Shuxin Song
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jingbo Wang
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaohu Ouyang
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Renyin Huang
- Jingshan Union Hospital, Union Hospital, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Fang Wang
- Jingshan Union Hospital, Union Hospital, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Junke Xie
- Jingshan Union Hospital, Union Hospital, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qianyun Chen
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Desheng Hu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- China-Russia Medical Research Center for Stress Immunology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
31
|
Luo Y, Li J, Fu Q, Zhang P, Song X, Liu M, Mo R, Fu J, Tang S, Wu J, Yang X, Liu X, Wang T, Ni G. Caerin 1.1 and 1.9 peptides induce acute caspase 3/GSDME-mediated pyroptosis in epithelial cancer cells. Sci Rep 2025; 15:13377. [PMID: 40251208 PMCID: PMC12008296 DOI: 10.1038/s41598-025-96438-0] [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: 08/14/2024] [Accepted: 03/28/2025] [Indexed: 04/20/2025] Open
Abstract
Caerin peptides exhibit a dual role in cancer treatment by directly killing cancer cells and modulating the tumour microenvironment to enhance anti-tumour immunity. This study investigates the mechanisms underlying caerin 1.1/1.9-induced acute cell death in epithelial cancer cells and explores their therapeutic potential. HeLa, A549, and Huh-7 cancer cell lines were treated with caerin 1.1/1.9 peptides. Morphological observations, flow cytometry, lactate dehydrogenase (LDH) release, and IL-18 secretion assays revealed the occurrence of pyroptosis following treatment. Specifically, a 1-h treatment with caerin 1.1/1.9 induced pyroptosis in HeLa, A549, and Huh-7 cells, characterised by cell swelling, membrane bubbling, and the release of IL-18 and LDH. Western blotting confirmed the upregulation of pyroptosis markers, including caspase-3, cleaved caspase-3, and GSDME-N fragments. These findings highlight the significant role of caerin peptides in inducing acute pyroptosis, a form of programmed cell death that enhances the immunogenicity of dying cancer cells, thus potentially improving the effectiveness of immunotherapies. This research underscores the therapeutic potential of caerin 1.1/1.9 peptides in cancer treatment, providing a foundation for developing new anti-cancer strategies that leverage both direct cytotoxic effects and immune modulation to achieve more effective and sustained anti-tumour responses.
Collapse
Affiliation(s)
- Yuandong Luo
- Medical College of Guizhou University, Guiyang, 550025, Guizhou, China
| | - Junjie Li
- Zhongao Biomedical Technology (Guangdong) Co. Ltd, Zhongshan, 528400, Guangdong, China
| | - Quanlan Fu
- Medical College of Guizhou University, Guiyang, 550025, Guizhou, China
| | - Pingping Zhang
- Cancer Research Institute, First People's Hospital of Foshan, Foshan, 528000, Guangdong, China
| | - Xinyi Song
- The First Affiliated Hospital/Clinical Medical School, Guangdong Pharmaceutical University, Guangzhou, 510080, China
| | - Mengqi Liu
- Medical College of Guizhou University, Guiyang, 550025, Guizhou, China
| | - Rongmi Mo
- The First Affiliated Hospital/Clinical Medical School, Guangdong Pharmaceutical University, Guangzhou, 510080, China
| | - Jiawei Fu
- The First Affiliated Hospital/Clinical Medical School, Guangdong Pharmaceutical University, Guangzhou, 510080, China
| | - Shuxian Tang
- Cancer Research Institute, First People's Hospital of Foshan, Foshan, 528000, Guangdong, China
| | - Jialing Wu
- Cancer Research Institute, First People's Hospital of Foshan, Foshan, 528000, Guangdong, China
| | - Xiaodan Yang
- The First Affiliated Hospital/Clinical Medical School, Guangdong Pharmaceutical University, Guangzhou, 510080, China
| | - Xiaosong Liu
- Medical College of Guizhou University, Guiyang, 550025, Guizhou, China.
- Cancer Research Institute, First People's Hospital of Foshan, Foshan, 528000, Guangdong, China.
| | - Tianfang Wang
- School of Science, Technology and Engineering, University of the Sunshine Coast, Maroochydore BC, QLD, 4558, Australia.
- Centre for Bioinnovation, University of the Sunshine Coast, Maroochydore BC, QLD, 4558, Australia.
| | - Guoying Ni
- Cancer Research Institute, First People's Hospital of Foshan, Foshan, 528000, Guangdong, China.
| |
Collapse
|
32
|
Zhang H, Tan B, Tang T, Tao J, Jin T, Wu S. Targeting inflammasomes as a therapeutic potential for HIV/AIDS. Cell Mol Life Sci 2025; 82:162. [PMID: 40244456 PMCID: PMC12006635 DOI: 10.1007/s00018-025-05685-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/24/2025] [Accepted: 03/29/2025] [Indexed: 04/18/2025]
Abstract
Human immunodeficiency virus (HIV) infection in humans can cause a variety of symptoms. Among these, acquired immunodeficiency syndrome (AIDS) remains the most severe form. Current treatment of HIV/AIDS with antiretroviral drugs effectively inhibits HIV replication and infection and significantly extends the lifespan of HIV/AIDS patients. However, antiretroviral drugs cannot completely remove HIV from patients due to the high latency of HIV, and they possess side effects and can lead to drug resistance. HIV/AIDS remains to be an incurable disease, and new methods and drugs are still desirable. Inflammasomes were found to be activated during HIV infection and regulate AIDS progression. Previous reviews provide a simple summary of inflammasome activators and inhibitors during HIV infection without distinguishing the specific infection stage, this kind of summary does not provide any clinical target value. Here, we provide a comprehensive review of inflammasomes in HIV/AIDS according to the infection timeline and propose several inflammasome target strategies for clinical HIV/AIDS treatment. We systematacially summarized the activation and function of kinds inflammasomes during the different HIV infection stages, with the aim of providing new therapeutic targets and directions for HIV/AIDS and HIV-associated comorbidities.
Collapse
Affiliation(s)
- Hongliang Zhang
- Center of Disease Immunity and Intervention, College of Medicine, Lishui University, Lishui, 323000, China
| | - Botao Tan
- Center of Disease Immunity and Intervention, College of Medicine, Lishui University, Lishui, 323000, China
| | - Tinbing Tang
- Center of Disease Immunity and Intervention, College of Medicine, Lishui University, Lishui, 323000, China
| | - Jinhui Tao
- Department of Rheumatology and Immunology, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, China.
| | - Tengchuan Jin
- Center of Disease Immunity and Intervention, College of Medicine, Lishui University, Lishui, 323000, China.
- Laboratory of Structural Immunology, CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China.
| | - Songquan Wu
- Center of Disease Immunity and Intervention, College of Medicine, Lishui University, Lishui, 323000, China.
| |
Collapse
|
33
|
Yang W, Xu Y, Tan Y, Lin J, Chen H, Li S, Miao H, Ye D. Molecular Mechanisms of Intervertebral Disc Degeneration Induced by Propionibacterium acnes. BIOMED RESEARCH INTERNATIONAL 2025; 2025:5513856. [PMID: 40264644 PMCID: PMC12014266 DOI: 10.1155/bmri/5513856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 03/14/2025] [Indexed: 04/24/2025]
Abstract
Intervertebral disc degeneration (IVDD), a prevalent degenerative disorder with substantial socioeconomic impacts, is closely linked to endplate inflammation and chronic low back pain. Its pathogenesis involves multifactorial mechanisms, including long-term chronic mechanical loading, external trauma, and hereditary factors. Emerging evidence highlights Propionibacterium acnes (P. acnes), a gram-positive bacterium with potent proinflammatory properties, as a key contributor to IVDD progression. This review systematically analyses the latest literature on related studies, focusing on the molecular mechanisms of IVDD induced by P. acnes. Three molecules play an important role in the induction of IVDD by P. acnes, namely, IL-1β, MIF, and MMP. In addition, P. acnes induces IVDD through three core mechanisms, namely, proinflammatory (activation of TLR2, production of large amounts of ROS to promote inflammation), pyroptosis (production of large amounts of NLRP3 through the TXNIP-NLRP3 axis and the ROS-NLRP3 axis), and apoptosis (promotion of Bax and inhibition of Bcl-2 expression through the TLR2-JNK pathway). The dissection of these related important molecules and pathogenic mechanisms can lead to a better understanding of the role of P. acnes in IVDD. It can provide an important theoretical basis for future research. However, the current study's lack of large-scale clinical validation, unresolved colonization controversies, and limited experimental methods are limitations. Therefore, in the future, it is still necessary to improve the relevant theories and resolve the current controversies through more advanced experimental methods and higher quality clinical studies. In conclusion, the study of P. acnes-induced IVDD is promising, and further research can be conducted in the future, which is expected to develop novel therapeutic approaches for P. acnes, thus effectively slowing down the development of IVDD.
Collapse
Affiliation(s)
- Weichao Yang
- Guangzhou Red Cross Hospital, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Yude Xu
- Department of Pain Medicine, Guangzhou Red Cross Hospital, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Yong Tan
- Guangzhou Red Cross Hospital, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Jinzhi Lin
- Guangzhou Red Cross Hospital, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Huan Chen
- Guangzhou Red Cross Hospital, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Shaojin Li
- Guangzhou Red Cross Hospital, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Haixiong Miao
- Guangzhou Red Cross Hospital, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| | - Dongping Ye
- Guangzhou Red Cross Hospital, Guangzhou Red Cross Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
34
|
Shi J, Yu Y, Yuan H, Li Y, Xue Y. Mitochondrial dysfunction in AMI: mechanisms and therapeutic perspectives. J Transl Med 2025; 23:418. [PMID: 40211347 PMCID: PMC11987341 DOI: 10.1186/s12967-025-06406-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 03/20/2025] [Indexed: 04/13/2025] Open
Abstract
Acute myocardial infarction (AMI) and the myocardial ischemia-reperfusion injury (MI/RI) that typically ensues represent a significant global health burden, accounting for a considerable number of deaths and disabilities. In the context of AMI, percutaneous coronary intervention (PCI) is the preferred treatment option for reducing acute ischemic damage to the heart. Despite the modernity of PCI therapy, pathological damage to cardiomyocytes due to MI/RI remains an important target for intervention that affects the long-term prognosis of patients. In recent years, mitochondrial dysfunction during AMI has been increasingly recognized as a critical factor in cardiomyocyte death. Damaged mitochondria play an active role in the formation of an inflammatory environment by triggering key signaling pathways, including those mediated by cyclic GMP-AMP synthase, NOD-like receptors and Toll-like receptors. This review emphasizes the dual role of mitochondria as both contributors to and regulators of inflammation. The aim is to explore the complex mechanisms of mitochondrial dysfunction in AMI and its profound impact on immune dysregulation. Specific interventions including mitochondrial-targeted antioxidants, membrane-stabilizing peptides, and mitochondrial transplantation therapies have demonstrated efficacy in preclinical AMI models.
Collapse
Affiliation(s)
- Jingle Shi
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yiding Yu
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Huajing Yuan
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yan Li
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China.
| | - Yitao Xue
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
35
|
Hou Y, Wang W, Ye J, Sun L, Zhou S, Zheng Q, Shi Y, Chen Y, Yao J, Wang L, Yan X, Wan R, Chen S, Li Y. The crucial role of neutrophil extracellular traps and IL-17 signaling in indomethacin-induced gastric injury in mice. Sci Rep 2025; 15:12109. [PMID: 40204883 PMCID: PMC11982219 DOI: 10.1038/s41598-025-95880-4] [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: 12/01/2024] [Accepted: 03/24/2025] [Indexed: 04/11/2025] Open
Abstract
The homeostasis of gastric mucosa is extremely delicate. Neutrophils, the most abundant immune cells in human circulation, are regarded crutial in the regulation of gastric mucosal immune response. Non-steroidal anti-inflammatory drugs (NSAIDs) induced gastric injury is the second major reason for gastric ulcers. The relations between neutrophils and Indomethacin-induced gastric injury are not fully understood. A mouse model of gastric injury was established using Indomethacin, followed by proteomic analysis (raw data are available via ProteomeXchange with identifier PXD058482). GO functional annotations and KEGG pathway enrichment analysis were conducted on significant differential proteins. The formation of neutrophil extracellular traps (NETs) was observed using ELISA and immunofluorescence. TEM, Western blot and Real-time PCR were applied to observe programmed death of gastric epithelial cells (GECs), and ELISA was conducted to measure levels of TNF-α and IL-1β in the gastric tissue. Deoxyribonuclease 1 (DNase 1), a NETs inhibitor, was administered intraperitoneally to inhibit NETs formation. In vitro, neutrophils were isolated from peripheral blood of mice and co-cultured with mouse GECs cell line, different dosage of Indomethacin were added to the culture dish, the levels of inflammatory factors, formation of NETs and GECs programmed death were assessed in vitro. Poly morphonuclear neutrophils (PMN) were extracted from mouse peripheral blood and single-cell RNA-sequencing (scRNA-seq) was further applied (raw data are available via Genome Sequence Archive with identifier CRA020950) to explore the intracellular mechanism of NETs formation. ELISA and immunofluorescence were performed to validate expression of IL-17 signaling pathway. After Indomethacin gavage, obvious gastric injury was observed. Proteomic analysis indicated that NETs formation played a crucial role in Indomethacin-induced gastric injury. Compared to control group, Indomethacin treatment resulted in NETs formation, elevated levels of TNF-α and IL-1β and GECs programmed death. Inhibition of NETs significantly reduced inflammatory factor levels and mitigated gastric injury caused by indomethacin. In vitro, 200 µL, 400 µL and 600 µL of Indomethacin caused excessive NETs formation in neutrophils. Besides, Indomethacin-induced NETs formation led to GECs programmed death in vitro. scRNA-seq revealed that neutrophils enrichment in the peripheral blood of Indomethacin-induced gastric injury and IL-17 signaling might be the key intracellular of NETs formation. Expressions of neutrophil IL-17R and concentration of IL-17 were significantly higher in model group. NETs formation is pivotal in Indomethacin-induced gastric injury, contributing to programmed cell death of GECs and inflammation; IL-17 signaling might be the key intracellular mechanism of NETs formation.
Collapse
Affiliation(s)
- Yujun Hou
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wen Wang
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiangnan Ye
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Luqiang Sun
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Siyuan Zhou
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qianhua Zheng
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yunzhou Shi
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ying Chen
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Junpeng Yao
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lu Wang
- Department of Acupuncture and Moxibustion, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiangyun Yan
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Renhong Wan
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shuai Chen
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ying Li
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
36
|
Wang L, Li HD, Sun X, Ni JH, Feng GZ, Shen XY, Weng HB, Fang H. The Protective Effects of Vanillic Acid on LPS-induced Acute Lung Injury by Inhibiting STIM1-mediated NLRP3 Inflammasome Activation. Inflammation 2025:10.1007/s10753-025-02293-6. [PMID: 40195181 DOI: 10.1007/s10753-025-02293-6] [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: 01/13/2025] [Accepted: 03/14/2025] [Indexed: 04/09/2025]
Abstract
Acute lung injury (ALI), which can progress to acute respiratory distress syndrome (ARDS), has inflammation as a crucial factor, especially the NOD-like receptor thermal protein domain associated protein 3 (NLRP3) inflammasome involvement. Stromal interaction molecule 1 (STIM1) can block NLRP3 activation, but the mechanism is unclear. Vanillic acid, possessing anti-inflammatory properties, has a role in acute lung injury (ALI) whose specific mechanism remains unclear. This study aimed to investigate the effectiveness of vanillic acid in ALI induced by lipopolysaccharides (LPS) and to elucidate the potential mechanisms. In vitro and in vivo experiments were conducted using cells and a mouse model to find out the impact and underlying mechanisms. We found that vanillic acid demonstrated significant inhibition of IL-1β and IL-18 release triggered by LPS and nigericin in J774A.1 cells. The in vivo findings indicated that vanillic acid not only mitigated acute lung injury but also suppressed NLRP3 inflammasome activation in mice. Mechanistically, vanillic acid inhibited the LPS-induced increase in STIM1 expression through the lysosomal degradation pathway. The reduced STIM1 expression diminished intracellular Ca2+ levels, thereby suppressing inflammasome activation and impeding the cleavage and maturation of Caspase-1 and GSDMD, and eventually attenuating cell pyroptosis. Vanillic acid exerts its inhibitory effects on NLRP3 inflammasome activation by promoting STIM1 degradation, thereby ameliorates ALI through impeding NLRP3-GSDMD mediated pyroptosis. The STIM1-NLRP3 signaling axis represents a promising avenue for potential therapeutic interventions in ALI.
Collapse
Affiliation(s)
- Lei Wang
- Department of Pharmacology, School of Pharmacy, Fudan University, No. 826, Zhangheng Road, Pudong New Area, Shanghai, 201203, China
| | - Hai-Dong Li
- Department of Pharmacology, School of Pharmacy, Fudan University, No. 826, Zhangheng Road, Pudong New Area, Shanghai, 201203, China
- Research and Translational Laboratory of Acute Injury and Secondary Infection, Minhang Hospital, Fudan University, Shanghai, China
| | - Xia Sun
- Department of Anesthesiology, Shanghai Geriatic Medical Center, Shanghai, 201104, China
| | - Jia-Hui Ni
- Department of Pharmacology, School of Pharmacy, Fudan University, No. 826, Zhangheng Road, Pudong New Area, Shanghai, 201203, China
| | - Gui-Ze Feng
- Department of Pharmacology, School of Pharmacy, Fudan University, No. 826, Zhangheng Road, Pudong New Area, Shanghai, 201203, China
| | - Xiao-Yan Shen
- Department of Pharmacology, School of Pharmacy, Fudan University, No. 826, Zhangheng Road, Pudong New Area, Shanghai, 201203, China
| | - Hong-Bo Weng
- Department of Pharmacology, School of Pharmacy, Fudan University, No. 826, Zhangheng Road, Pudong New Area, Shanghai, 201203, China.
| | - Hao Fang
- Department of Anesthesiology, Shanghai Geriatic Medical Center, Shanghai, 201104, China.
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
37
|
Ma X, Lin Y, Zhang L, Huang Z, Zhang Y, Fu X, Li P. The dual missions of FoxO3a in inflammatory diseases: Regulation of antioxidant enzymes and involvement in programmed cell death. Int Immunopharmacol 2025; 151:114369. [PMID: 40031428 DOI: 10.1016/j.intimp.2025.114369] [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: 01/20/2025] [Revised: 02/22/2025] [Accepted: 02/23/2025] [Indexed: 03/05/2025]
Abstract
The transcription factor FoxO3a plays a crucial role in the process of cells adapting to various stress conditions. Multiple post - translational modifications and epigenetic mechanisms work together to precisely regulate the activity of FoxO3a, influencing its subcellular localization, stability, interactions with other proteins, DNA - binding affinity, and transcriptional regulatory capacity. Under different chemical signal stimuli and subcellular environments, the activation of FoxO3a triggered by oxidative stress can initiate diverse transcriptional programs, which are essential for the body to resist oxidative damage. In the development and progression of inflammatory diseases, FoxO3a exerts an important function by regulating the expression levels of antioxidant enzymes and participating in key physiological processes such as programmed cell death. This article comprehensively reviews the structural characteristics, mechanism of action of FoxO3a, as well as its functions in regulating antioxidant enzymes and programmed cell death. The aim is to deeply explore the potential of FoxO3a as a potential therapeutic target for preventing and treating damages such as inflammatory diseases caused by cellular stress.
Collapse
Affiliation(s)
- Xiangli Ma
- Department of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Yujie Lin
- Department of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Ling Zhang
- Department of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Zhenzhen Huang
- Department of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Yurong Zhang
- Department of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Xu Fu
- Key Laboratory of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Peiwu Li
- Department of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, China.
| |
Collapse
|
38
|
Ding Y, Liu Y, Li D, Hu R, Tian Z, Yang L, Li Y, Lin Y, Qu Y. Melatonin Ameliorates Senescence of Mouse Auditory Cell Line HEI-OC1 Cells by Suppressing NLRP3 Inflammasome-Mediated Pyroptosis. Mol Neurobiol 2025:10.1007/s12035-025-04880-y. [PMID: 40169516 DOI: 10.1007/s12035-025-04880-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 03/21/2025] [Indexed: 04/03/2025]
Abstract
The aim of this study was to determine whether oxidative stress-induced premature senescence in mouse auditory cell line HEI-OC1 cells in vitro is associated with NLRP3 inflammasome activation and pyroptosis, and whether melatonin has a protective effect. HEI-OC1 cells were exposed to different concentrations of hydrogen peroxide (H2O2) to induce oxidative stress and subsequently analyzed by Western blotting to measure pyroptosis-related proteins - NLRP3, caspase-1, and GSDMD-N. Compared with untreated control cells, exposure to different concentrations of hydrogen peroxide (H2O2) promoted premature senescence of HEI-OC1 cells, accompanied by a significant increase in levels of pyroptosis-related proteins - NLRP3, caspase-1, and GSDMD-N. Furthermore, melatonin treatment was shown to decrease the expression of these proteins in HEI-OC1 cells and attenuate the H2O2-induced senescence process. NLRP3 inflammasome activation contributes to oxidative stress-induced premature senescence of HEI-OC1 cells in vitro, leading to pyroptosis. Melatonin attenuates pyroptosis and senescence in HEI-OC1 cells by inhibiting the expression of NLRP3, caspase-1, and GSDMD-N, providing reliable evidence for melatonin as a potential therapeutic agent for age-related hearing loss.
Collapse
Affiliation(s)
- Yongqing Ding
- Department of Otolaryngology, Qiaoxi District, The Third Hospital of Hebei Medical University, No. 13, Ziqiang Road, Shijiazhuang City, Hebei Province, China
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Yachao Liu
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Dong Li
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Ruili Hu
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Zedong Tian
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Lihang Yang
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Yanping Li
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Yantao Lin
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Yan Qu
- Department of Otolaryngology, Qiaoxi District, The Third Hospital of Hebei Medical University, No. 13, Ziqiang Road, Shijiazhuang City, Hebei Province, China.
| |
Collapse
|
39
|
Wang M, Tu T, Wang Y, Tian L, Yang Y. Salidroside alleviates imiquimod-induced psoriasis by inhibiting GSDMD-driven keratinocyte pyroptosis. Biotechnol Appl Biochem 2025; 72:355-368. [PMID: 39279255 DOI: 10.1002/bab.2668] [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: 04/15/2024] [Accepted: 08/31/2024] [Indexed: 09/18/2024]
Abstract
Psoriasis is a common immune-related polygenic inflammatory skin disease. Salidroside (SAL) exerts anti-inflammatory and antioxidant effects and is used to treat skin diseases. However, the specific effects of SAL on psoriasis remain unclear. In this study, we aimed to investigate the efficacy of SAL for psoriasis treatment. Mice were treated with imiquimod (IMQ) to establish an in vivo psoriasis model. Histological analysis was conducted via hematoxylin and eosin staining. Cytokine release was determined via enzyme-linked immunosorbent assay. Additionally, mRNA levels were determined via reverse transcription-quantitative polymerase chain reaction. Protein expression was assessed via Western blotting. Gasdermin D (GSDMD) and Ki-67 expression levels were determined via immunohistochemistry. Caspase 1 and GSDMD expression levels were determined via immunofluorescence assay. Furthermore, macrophage function and keratinocyte pyroptosis were also analyzed via flow cytometry. Cell proliferation was determined using 5-ethynyl-2'deoxyuridine assay. SAL alleviated IMQ-induced psoriasis. IMQ-mediated GSDMD-driven pyroptosis and keratinocyte hyperproliferation promoted M1 macrophage polarization. However, SAL treatment suppressed GSDMD expression, thereby inhibiting keratinocyte proliferation and pyroptosis and promoting M2 macrophage polarization. GSDMD deficiency further promoted the effects of SAL and suppressed psoriasis progression. Overall, our findings suggest that SAL exerts protective effects against psoriasis. Specifically, it exerts anti-inflammatory effects by regulating M2 macrophage polarization and inhibiting keratinocyte pyroptosis-driven proliferation induced by the immune microenvironment in psoriasis.
Collapse
Affiliation(s)
- Mengjie Wang
- Department of Dermatology and Surgery, Baotou Medical College of Inner Mongolia University of Science and Technology, Baotou, China
| | - Tuyagaer Tu
- Department of Dermatology and Surgery, Baotou Medical College of Inner Mongolia University of Science and Technology, Baotou, China
| | - Yangxingyun Wang
- Department of Dermatology and Surgery, Baotou Medical College of Inner Mongolia University of Science and Technology, Baotou, China
| | - Limin Tian
- Department of Dermatology, The First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, China
| | - Yuenan Yang
- Department of Dermatology, The First Affiliated Hospital of Baotou Medical College, Inner Mongolia University of Science and Technology, Baotou, China
| |
Collapse
|
40
|
Tian L, Piao S, Li X, Guo L, Huang L, Gao W. Functional Materials Targeted Regulation of Gasdermins: From Fundamentals to Functionalities and Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2500873. [PMID: 40273335 PMCID: PMC12021126 DOI: 10.1002/advs.202500873] [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: 01/14/2025] [Revised: 02/19/2025] [Indexed: 04/26/2025]
Abstract
Targeted regulation of pyroptosis to modulate the immune landscape has emerged as a novel design strategy for cancer immunotherapy and anti-inflammatory therapy. However, pyroptosis acts as a double-edged sword, making it important to optimize the design strategies of functional materials to appropriately activate pyroptosis for effective disease treatment. This paper summarizes and discusses the structure, pore formation, and molecular mechanisms of "executor" Gasdermins, as well as the events preceding and following these processes. Subsequently, the focus is on reviewing functional materials that directly regulate Gasdermin pore formation to target pyroptosis and those that indirectly regulate the events before and after Gasdermin pore formation to control pyroptosis activity. Finally, the advantages, disadvantages, and future prospects of designing such functional materials are provided, aiming to facilitate the precise design, pharmacological investigation, and clinical translation of pyroptosis-related functional materials.
Collapse
Affiliation(s)
- Luyao Tian
- School of Pharmaceutical Science and TechnologyTianjin UniversityTianjin300072P. R. China
| | - Shuo Piao
- School of Pharmaceutical Science and TechnologyTianjin UniversityTianjin300072P. R. China
| | - Xia Li
- School of Pharmaceutical Science and TechnologyTianjin UniversityTianjin300072P. R. China
| | - Lanping Guo
- National Resource Center for Chinese Materia MedicaChina Academy of Chinese Medical SciencesBeijing100700P. R. China
| | - Luqi Huang
- National Resource Center for Chinese Materia MedicaChina Academy of Chinese Medical SciencesBeijing100700P. R. China
| | - Wenyuan Gao
- Key Laboratory of Pharmacology School of Pharmaceutical Science and TechnologyTianjin UniversityTianjin300072P. R. China
| |
Collapse
|
41
|
Xu W, Huang Y, Zhou R. NLRP3 inflammasome in neuroinflammation and central nervous system diseases. Cell Mol Immunol 2025; 22:341-355. [PMID: 40075143 PMCID: PMC11955557 DOI: 10.1038/s41423-025-01275-w] [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: 12/06/2024] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
Neuroinflammation plays an important role in the pathogenesis of various central nervous system (CNS) diseases. The NLRP3 inflammasome is an important intracellular multiprotein complex composed of the innate immune receptor NLRP3, the adaptor protein ASC, and the protease caspase-1. The activation of the NLRP3 inflammasome can induce pyroptosis and the release of the proinflammatory cytokines IL-1β and IL-18, thus playing a central role in immune and inflammatory responses. Recent studies have revealed that the NLRP3 inflammasome is activated in the brain to induce neuroinflammation, leading to further neuronal damage and functional impairment, and contributes to the pathological process of various neurological diseases, such as multiple sclerosis, Parkinson's disease, Alzheimer's disease, and stroke. In this review, we summarize the important role of the NLRP3 inflammasome in the pathogenesis of neuroinflammation and the pathological course of CNS diseases and discuss potential approaches to target the NLRP3 inflammasome for the treatment of CNS diseases.
Collapse
Grants
- 81821001, 82130107, 82330052, 82202038, U20A20359 National Natural Science Foundation of China (National Science Foundation of China)
- National Key research and development program of China (grant number (2020YFA0509101), The Strategic Priority Research Program of the Chinese Academy of Sciences (XDB0940000),
- MEXT | JST | Strategic Promotion of Innovative R and D (Strategic Promotion of Innovative R&D)
- the CAS Project for Young Scientists in Basic Research (YSBR-074) and the Fundamental Research Funds for the Central Universities, the outstanding Youth Project of Anhui Provincial Natural Science Foundation (2408085Y049), the Research Start-up Funding of the Institute of Health and Medicine, Hefei Comprehensive National Science Center (2024KYQD004), the Natural Science Foundation of Jiangsu Province (BK20221085),
- The key project of Anhui Provincial Department of Education Fund (2024AH052060).
Collapse
Affiliation(s)
- Wen Xu
- Neurology Department, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, P. R. China
| | - Yi Huang
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, 230601, China.
| | - Rongbin Zhou
- National Key Laboratory of Immune Response and Immunotherapy, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230027, China.
- Department of Geriatrics, Gerontology Institute of Anhui Province, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China.
| |
Collapse
|
42
|
Chi X, Ding J, Zhang Y, Chen Y, Han Y, Lin Y, Jiang J. Berberine protects against dysentery by targeting both Shigella filamentous temperature sensitive protein Z and host pyroptosis: Resolving in vitro-vivo effect discrepancy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156517. [PMID: 39986228 DOI: 10.1016/j.phymed.2025.156517] [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: 10/06/2024] [Revised: 01/29/2025] [Accepted: 02/13/2025] [Indexed: 02/24/2025]
Abstract
BACKGROUND Berberine (BBR), an isoquinoline alkaloid, has been applied clinically to treat dysentery caused by Shigella for decades. Nevertheless, the precise mechanisms behind its anti-Shigella effect have not been fully elucidated. PURPOSE This study aims to investigate the mechanism of BBR on antibacterial activity against S. flexneri infection. METHODS We initially reproduced the mouse model of Shigella flexneri-induced dysentery, and then, assessed the therapeutic effect of BBR. In vitro, we measured the inhibitory effect of BBR against S. flexneri and the GTPase activity of FtsZ (filamentous temperature sensitive protein Z) using the minimum inhibitory concentration (MIC) test and an enzyme activity assay to investigate the bacteria-directed mechanisms. Subsequently, we utilized both the in vivo mouse model of dysentery and the in vitro macrophage infection model with S. flexneri to explore the host-directed anti-Shigella mechanisms of BBR. The canonical pyroptosis pathway mediated by caspase-1 and mitochondrial damage were examined by Western blot, immunofluorescence and RNA interference analysis. RESULTS Administration of BBR alleviated the symptoms of dysentery induced by S. flexneri infection. In vitro, BBR could inhibit the growth of S. flexneri by targeting the GTPase activity of FtsZ, thereby affecting bacterial cell division. Additionally, our in vivo findings revealed that BBR suppressed macrophage pyroptosis by inhibiting the expression of caspase-1 and subsequently the mitochondrial damage, which in turn reduced the intestinal inflammation and tissue damage. CONCLUSIONS Our results provide a novel mechanism of BBR's action, which targets both the bacterium and the host to exert its antibacterial effects. Furthermore, it also provides an explanation for the discrepancy between BBR's relatively modest antibacterial efficacy in vitro and its enhanced antibacterial effects in vivo, thus, giving support to its clinical use.
Collapse
Affiliation(s)
- Xiangyin Chi
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Jinwen Ding
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yu Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Ying Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yanxing Han
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yuan Lin
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Jiandong Jiang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
43
|
Luo QY, Yang J, Di T, Xia ZF, Zhang L, Pan WT, Shi S, Yang LQ, Sun J, Qiu MZ, Yang DJ. The novel BCL-2/BCL-XL inhibitor APG-1252-mediated cleavage of GSDME enhances the antitumor efficacy of HER2-targeted therapy in HER2-positive gastric cancer. Acta Pharmacol Sin 2025; 46:1082-1096. [PMID: 39592733 PMCID: PMC11950313 DOI: 10.1038/s41401-024-01414-5] [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] [Received: 06/21/2024] [Accepted: 10/20/2024] [Indexed: 11/28/2024]
Abstract
HER2-positive gastric cancer has a poor prognosis, with a high incidence of drug resistance and a lack of effective treatments for drug-resistant patients. The exploration of the mechanism of resistance to HER2-targeted therapy in HER2-positive gastric cancer and the identification of effective strategies to reverse it are urgently needed. In this study, we found that HER2-targeted agents upregulated the expression of GSDME and that the overexpression of GSDME attenuated the sensitivity of HER2-targeted agents. Furthermore, we observed that the BCL-2/BCL-XL inhibitor APG-1252 plus lapatinib promoted GSDME-mediated pyroptosis and exhibited remarkable antitumor activity both in vitro and in vivo. Mechanistically, APG-1252 combined with lapatinib synergistically induced GSDME-mediated pyroptosis in HER2-positive gastric cancer by activating caspase-dependent pathways and blocking the phospho-AKT/GSK-3β/MCL-1 signaling pathway. Our data indicated that the combination of lapatinib and APG-1252 had a synergistic antitumor effect on HER2-positive gastric cancer through the induction of caspase-3/GSDME-mediated apoptosis and pyroptosis.
Collapse
Affiliation(s)
- Qiu-Yun Luo
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
- Department of Clinical Research Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510060, China
| | - Jing Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Tian Di
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Zeng-Fei Xia
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Lin Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
- Department of Clinical Laboratory, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Wen-Tao Pan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
- Ascentage Pharma (Suzhou) Co, Ltd, Suzhou, 215000, China
| | - Shan Shi
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Li-Qiong Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Jian Sun
- Department of Clinical Research Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510060, China.
| | - Miao-Zhen Qiu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| | - Da-Jun Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
- Ascentage Pharma (Suzhou) Co, Ltd, Suzhou, 215000, China.
| |
Collapse
|
44
|
Lin M, Zhang C, Li H, Li K, Gou S, He X, Lv C, Gao K. Pyroptosis for osteoarthritis treatment: insights into cellular and molecular interactions inflammatory. Front Immunol 2025; 16:1556990. [PMID: 40236711 PMCID: PMC11996656 DOI: 10.3389/fimmu.2025.1556990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 03/13/2025] [Indexed: 04/17/2025] Open
Abstract
Osteoarthritis (OA) is a widely prevalent chronic degenerative disease often associated with significant pain and disability. It is characterized by the deterioration of cartilage and the extracellular matrix (ECM), synovial inflammation, and subchondral bone remodeling. Recent studies have highlighted pyroptosis-a form of programmed cell death triggered by the inflammasome-as a key factor in sustaining chronic inflammation. Central to this process are the inflammatory cytokines interleukin-1β (IL-1β) and interleukin-18 (IL-18), which play crucial roles mediating intra-articular pyroptosis through the NOD-like receptor protein 3 (NLRP3) inflammasome. This paper investigates the role of the pyroptosis pathway in perpetuating chronic inflammatory diseases and its linkage with OA. Furthermore, it explores the mechanisms of pyroptosis, mediated by nuclear factor κB (NF-κB), the purinergic receptor P2X ligand-gated ion channel 7 (P2X7R), adenosine monophosphate (AMP)-activated protein kinase (AMPK), and hypoxia-inducible factor-1α (HIF-1α). Additionally, it examines the interactions among various cellular components in the context of OA. These insights indicate that targeting the regulation of pyroptosis presents a promising therapeutic approach for the prevention and treatment of OA, offering valuable theoretical perspectives for its effective management.
Collapse
Affiliation(s)
- Minghui Lin
- Second College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Cunxin Zhang
- Department of Orthopedics, Jining No.1 People’s Hospital, Jining, China
| | - Haiming Li
- Second College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Kang Li
- Department of Orthopedics, Jining No.1 People’s Hospital, Jining, China
| | - Shuao Gou
- Jining No.1 People's Hospital, affiliated with Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xiao He
- Department of Orthopedics, Jining No.1 People’s Hospital, Jining, China
- Medical Integration and Practice Center, Shandong University, Jinan, China
| | - Chaoliang Lv
- Department of Orthopedics, Jining No.1 People’s Hospital, Jining, China
| | - Kai Gao
- Department of Orthopedics, Jining No.1 People’s Hospital, Jining, China
| |
Collapse
|
45
|
Gao H, Xie T, Li Y, Xu Z, Song Z, Yu H, Zhou H, Li W, Yun C, Guan B, Luan S, Yin L. Role of gasdermins in chronic kidney disease. Front Immunol 2025; 16:1557707. [PMID: 40236694 PMCID: PMC11996640 DOI: 10.3389/fimmu.2025.1557707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/14/2025] [Indexed: 04/17/2025] Open
Abstract
Gasdermins (GSDMs), functioning as membrane perforating proteins, can be activated by canonical inflammasomes, noncanonical inflammasomes, as well as non-inflammasomes, leading to cell pyroptosis and the subsequent release of inflammatory mediators. Increasing evidence has implicated that GSDMs are associated with chronic kidney disease (CKD), including diabetes nephropathy, lupus nephritis, obstructive nephropathy, and crystalline nephropathy. This review centers on the role of GSDMs-mediated pyroptosis in the pathogenesis of CKD, providing novel ideas for enhancing the prognosis and therapeutic strategies of CKD.
Collapse
Affiliation(s)
- Hanchao Gao
- Department of Nephrology, Shenzhen Longhua District Central Hospital, Shenzhen Longhua District Key Laboratory for Diagnosis and Treatment of Chronic Kidney Disease, Shenzhen, Guangdong, China
| | - Ting Xie
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Yunyi Li
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Zigan Xu
- Department of Nephrology, Shenzhen Longhua District Central Hospital, Shenzhen Longhua District Key Laboratory for Diagnosis and Treatment of Chronic Kidney Disease, Shenzhen, Guangdong, China
| | - Zhuoheng Song
- Department of Nephrology, Shenzhen Longhua District Central Hospital, Shenzhen Longhua District Key Laboratory for Diagnosis and Treatment of Chronic Kidney Disease, Shenzhen, Guangdong, China
| | - Huixia Yu
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Hongming Zhou
- Department of Nephrology, Shenzhen Longhua District Central Hospital, Shenzhen Longhua District Key Laboratory for Diagnosis and Treatment of Chronic Kidney Disease, Shenzhen, Guangdong, China
| | - Weilong Li
- Department of Nephrology, Shenzhen Longhua District Central Hospital, Shenzhen Longhua District Key Laboratory for Diagnosis and Treatment of Chronic Kidney Disease, Shenzhen, Guangdong, China
| | - Chen Yun
- Charité-Universitätsmedizin Berlin, Campus Mitte, Berlin, Germany
| | - Baozhang Guan
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Shaodong Luan
- Department of Nephrology, Shenzhen Longhua District Central Hospital, Shenzhen Longhua District Key Laboratory for Diagnosis and Treatment of Chronic Kidney Disease, Shenzhen, Guangdong, China
| | - Lianghong Yin
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| |
Collapse
|
46
|
Zheng YY, Shen DN, Peng XL, San WQ, Zhou QY, Yang SJ, Meng GL, Shi JH, Chen Y. TRADD-mediated pyroptosis contributes to diabetic cardiomyopathy. Acta Pharmacol Sin 2025; 46:940-950. [PMID: 39753984 PMCID: PMC11950311 DOI: 10.1038/s41401-024-01450-1] [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] [Received: 06/24/2024] [Accepted: 11/29/2024] [Indexed: 03/17/2025]
Abstract
Regulated cell death like pyroptosis is one vital cause of diabetic cardiomyopathy (DCM), which eventually leads to heart failure. Tumor necrosis factor (TNF) receptor-associated death domain protein (TRADD) is an adapter protein with multiple functions that participates in the pathophysiological progress of different cardiovascular disorders via regulating regulated cell death. Studies have shown that TRADD combines with receptor-interacting protein kinase 3 (RIPK3) and facilitates its activation, thereby mediating TNF-induced necroptosis. However, no direct relationship between TRADD and pyroptosis has been identified. In this study, we investigated the role and mechanisms of TRADD in pyroptosis during DCM. We established a streptozotocin (STZ)-induced diabetic mouse model and high glucose (HG)-treated cardiomyocytes model. We showed that the expression levels of TRADD were significantly increased in the hearts of diabetic mice and HG-treated cardiomyocytes. Knockdown of TRADD did not affect blood glucose and triglyceride levels, but significantly improved cardiac function, and attenuated myocardial hypertrophy, fibrosis, and pyroptosis in the heart of diabetic mice. Furthermore, both knockdown of TRADD and application of TRADD inhibitor apostatin-1 (Apt-1, 10 μM) significantly ameliorated cell injury and pyroptosis in HG-treated cardiomyocytes. We demonstrated that HG treatment increased the expression of X-box binding protein 1 (XBP1) and enhanced the binding of XBP1 to the TRADD promoter to elevate TRADD expression in the cardiomyocytes. Collectively, this study provides evidence that TRADD-mediated pyroptosis contributes to DCM, suggesting that strategies to inhibit TRADD activity may be a novel approach for DCM treatment.
Collapse
Affiliation(s)
- Yang-Yang Zheng
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, 226001, China
- Department of Pharmacy, Xuzhou Oriental Hospital Affiliated to Xuzhou Medical University, Xuzhou, 221004, China
| | - Dan-Ning Shen
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, 226001, China
| | - Xiao-Lu Peng
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, 226001, China
| | - Wen-Qing San
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, 226001, China
| | - Qian-You Zhou
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, 226001, China
| | - Sheng-Ju Yang
- School of Medicine, Nantong University, Nantong, 226001, China
| | - Guo-Liang Meng
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, 226001, China.
| | - Jia-Hai Shi
- Department of Thoracic Surgery, Affiliated Hospital of Nantong University, Nantong, 226006, China.
| | - Yun Chen
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, 226001, China.
| |
Collapse
|
47
|
Mei Y, Chen X, Shi S, Lin W, Cheng Z, Fan X, Wu W, Han J, Huang W, Ye B, Dai S. GI-Y2, a novel gasdermin D inhibitor, attenuates sepsis-induced myocardial dysfunction by inhibiting gasdermin D-mediated pyroptosis in macrophages. Br J Pharmacol 2025. [PMID: 40165368 DOI: 10.1111/bph.70040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 03/01/2025] [Accepted: 03/04/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND AND PURPOSE Myocardial dysfunction is a significant complication associated with sepsis. However, there are currently no specific and effective treatments available. Inhibiting gasdermin D (GSDMD)-mediated pyroptosis has shown promise in mitigating sepsis-induced myocardial dysfunction. The GSDMD inhibitor Y2 (GI-Y2) has been demonstrated to directly bind to GSDMD. Nonetheless, it remains uncertain whether GI-Y2 offers a cardioprotective effect in the context of sepsis-induced myocardial dysfunction. EXPERIMENTAL APPROACH A mouse model of sepsis was created using lipopolysaccharide (LPS), caecal ligation and puncture. Following treatment with GI-Y2 or macrophage membrane-encapsulated GI-Y2 nanoparticles (GI-Y2@MM-NPs), myocardial dysfunction and pyroptosis levels in heart tissues were assessed. Transcriptome sequencing revealed the molecular mechanism of GI-Y2 in treating septic cardiomyopathy. KEY RESULTS We observed that GI-Y2 alleviated myocardial dysfunction and attenuated cardiac inflammation in mice induced by LPS, caecal ligation and puncture. GI-Y2 reduced macrophage pyroptosis and attenuated macrophage-mediated cardiomyocyte injury induced by LPS/nigericin. Concurrently, we confirmed the protective effect of GI-Y2 against LPS-induced cardiac dysfunction was abolished in the absence of GSDMD. Additionally, GI-Y2 attenuated the mitochondrial damage induced by LPS by inhibiting GSDMD in the mitochondria. Furthermore, we developed GI-Y2@MM-NPs to enhance the targeting capability of GI-Y2 towards macrophages in heart tissues and demonstrated its protective effect in vivo. CONCLUSION AND IMPLICATIONS These findings indicate that GI-Y2 alleviates septic myocardial injury and dysfunction by specifically targeting GSDMD, thereby inhibiting GSDMD-mediated pyroptosis and mitochondrial damage. Both GI-Y2 and GI-Y2@MM-NPs may serve as promising therapeutic options for addressing septic myocardial dysfunction.
Collapse
Affiliation(s)
- Yiling Mei
- The Key Laboratory of Emergency and Disaster Medicine of Wenzhou, Department of Emergency, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xudong Chen
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Cardiology, Ningbo Hangzhou Bay Hospital, Ningbo, Zhejiang, China
| | - Si Shi
- First School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wante Lin
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhenfeng Cheng
- Huzhou Central Hospital, Affiliated Central Hospital of Huzhou University, Huzhou, Zhejiang, China
| | - Xiaoxi Fan
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wenqi Wu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jibo Han
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| | - Weijian Huang
- The Key Laboratory of Emergency and Disaster Medicine of Wenzhou, Department of Emergency, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Bozhi Ye
- Department of Cardiology and The Key Laboratory of Cardiovascular Disease of Wenzhou, the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shanshan Dai
- The Key Laboratory of Emergency and Disaster Medicine of Wenzhou, Department of Emergency, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
48
|
Wen X, Fan J, Duan X, Zhu X, Bai J, Zhang T. Mitochondrial DNA in Exercise-Mediated Innate Immune Responses. Int J Mol Sci 2025; 26:3069. [PMID: 40243714 PMCID: PMC11988935 DOI: 10.3390/ijms26073069] [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: 03/07/2025] [Revised: 03/25/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Abstract
Mitochondria are considered as "the plant of power" with cells for a long time. However, recent researches suggest that mitochondria also take part in innate immune response to a great extent. Remarkably, mtDNA was reported to have immunnostimulatory potential in 2004. Since then, there has been rapid growth in understanding the role of mtDNA in innate immune. The mtDNA is released into cytosol, extracellular environment, or circulating blood through BAK/BAX pore, mPTP, and GSDMD pore upon mitochondrial damage, where it is recognized by PRRs including TLR9, cGAS, and NLRP3, thereby triggering innate immune response. On the other hand, regular exercise has been recognized as an effective intervention strategy for innate immune response. Some studies show that chronic moderate-intensity endurance exercise, resistance training, HIIT, and moderate-intensity acute exercise enhance mitochondrial function by promoting mtDNA transcription and replication, thus blunting the abnormal release of mtDNA and excessive innate immune response. On the contrary, high-intensity acute exercise elicits the opposite effect. Nevertheless, only a very small body of research by far has been performed to illustrate the impact of exercise on mtDNA-driven innate immune response, and an overall review is lacking. In light of these, we summarize the current knowledge on the mechanism mediating the release of mtDNA, the role of mtDNA in innate immune response and the influence of exercise on mtDNA leakage, hoping to pave the way to investigate new diagnostic and therapeutic approaches for immunopathies.
Collapse
Affiliation(s)
| | | | | | | | | | - Tan Zhang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China
| |
Collapse
|
49
|
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
|
50
|
He S, Huang Q, Cheng J. The conflicting role highlights the complexity of GSDMs in cancer. Front Immunol 2025; 16:1531695. [PMID: 40201182 PMCID: PMC11975587 DOI: 10.3389/fimmu.2025.1531695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 03/10/2025] [Indexed: 04/10/2025] Open
Abstract
Gasdermins (GSDMs) are an important family of proteins that have received extensive attention in tumor research in recent years. They directly induce tumor cell death by mediating pyroptosis and also regulate the recognition and clearance of tumor cells by the immune system by affecting the microenvironment. Therefore, it is of great significance to investigate the role of GSDMs in tumor development and tumor microenvironment. It can not only reveal new mechanisms of cancer development, but also provide theoretical basis for the development of novel anti-tumor therapeutic strategies. This literature review aims to systematically summarize the dual roles of GSDMs in tumor development and their interactions with the tumor microenvironment, and to focus on the importance of GSDM-mediated pyroptosis in anti-cancer therapy, with a view to providing guidance for future research directions.
Collapse
Affiliation(s)
- Sijia He
- Cancer Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Oncology, Jiuquan Branch of Shanghai General Hospital, Jiuquan, Gansu, China
| | - Qian Huang
- Cancer Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jin Cheng
- Cancer Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|