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
|
Fang J, Li Z, Wang P, Zhang X, Mao S, Li Y, Yu D, Li X, Xing Y, Shi H, Yin S. Inhibition of the NLRP3 inflammasome attenuates spiral ganglion neuron degeneration in aminoglycoside-induced hearing loss. Neural Regen Res 2025; 20:3025-3039. [PMID: 39610108 PMCID: PMC11826467 DOI: 10.4103/nrr.nrr-d-23-01879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 02/08/2024] [Accepted: 03/28/2024] [Indexed: 11/30/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202510000-00031/figure1/v/2024-11-26T163120Z/r/image-tiff Aminoglycosides are a widely used class of antibacterials renowned for their effectiveness and broad antimicrobial spectrum. However, their use leads to irreversible hearing damage by causing apoptosis of hair cells as their direct target. In addition, the hearing damage caused by aminoglycosides involves damage of spiral ganglion neurons upon exposure. To investigate the mechanisms underlying spiral ganglion neuron degeneration induced by aminoglycosides, we used a C57BL/6J mouse model treated with kanamycin. We found that the mice exhibited auditory deficits following the acute loss of outer hair cells. Spiral ganglion neurons displayed hallmarks of pyroptosis and exhibited progressive degeneration over time. Transcriptomic profiling of these neurons showed significant upregulation of genes associated with inflammation and immune response, particularly those related to the NLRP3 inflammasome. Activation of the canonical pyroptotic pathway in spiral ganglion neurons was observed, accompanied by infiltration of macrophages and the release of proinflammatory cytokines. Pharmacological intervention targeting NLRP3 using Mcc950 and genetic intervention using NLRP3 knockout ameliorated spiral ganglion neuron degeneration in the injury model. These findings suggest that NLRP3 inflammasome-mediated pyroptosis plays a role in aminoglycoside-induced spiral ganglion neuron degeneration. Inhibition of this pathway may offer a potential therapeutic strategy for treating sensorineural hearing loss by reducing spiral ganglion neuron degeneration.
Collapse
Affiliation(s)
- Jia Fang
- Department of Otolaryngology Head and Neck Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine; Otolaryngology Institute of Shanghai Jiao Tong University; Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
- Department of Otorhinolaryngology-Head and Neck Surgery, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhuangzhuang Li
- Department of Otolaryngology Head and Neck Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine; Otolaryngology Institute of Shanghai Jiao Tong University; Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
| | - Pengjun Wang
- Department of Otolaryngology Head and Neck Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine; Otolaryngology Institute of Shanghai Jiao Tong University; Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
| | - Xiaoxu Zhang
- Department of Otolaryngology Head and Neck Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine; Otolaryngology Institute of Shanghai Jiao Tong University; Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
| | - Song Mao
- Department of Otolaryngology Head and Neck Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine; Otolaryngology Institute of Shanghai Jiao Tong University; Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
| | - Yini Li
- Department of Otolaryngology Head and Neck Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine; Otolaryngology Institute of Shanghai Jiao Tong University; Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
| | - Dongzhen Yu
- Department of Otolaryngology Head and Neck Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine; Otolaryngology Institute of Shanghai Jiao Tong University; Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
| | - Xiaoyan Li
- Department of Otorhinolaryngology-Head and Neck Surgery, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yazhi Xing
- Department of Otolaryngology Head and Neck Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine; Otolaryngology Institute of Shanghai Jiao Tong University; Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
| | - Haibo Shi
- Department of Otolaryngology Head and Neck Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine; Otolaryngology Institute of Shanghai Jiao Tong University; Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
| | - Shankai Yin
- Department of Otolaryngology Head and Neck Surgery, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine; Otolaryngology Institute of Shanghai Jiao Tong University; Shanghai Key Laboratory of Sleep Disordered Breathing, Shanghai, China
| |
Collapse
|
3
|
Cui Y, Zhang M, Xiao Y, Liu J, Chen Y, Ruan X, Zhao X, Liu Y, Shi Y, Tian J, Li L, Zhang X, Jia M, Wang Y, Yang X, Lin Z, Chen J. MLKL triggers NLRP3 activation in sodium arsenite-induced myocardial necroinflammation. Toxicology 2025; 515:154132. [PMID: 40188932 DOI: 10.1016/j.tox.2025.154132] [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: 02/20/2025] [Revised: 03/31/2025] [Accepted: 04/02/2025] [Indexed: 04/13/2025]
Abstract
Prolonged exposure to arsenic elevates the risk of developing a range of cardiovascular disorders. However, the mechanisms underlying myocardial damage from arsenic exposure remain elusive. Our earlier research suggest that drinking arsenic-contaminated water can lead to substantial inflammatory and necrotic injury in the myocardium of rats. This study was to ascertain whether mixed lineage kinase domain-like protein (MLKL) triggers Nod-like receptor protein-3 (NLRP3) activation during arsenic-induced myocardial necroinflammation in H9C2 cardiomyocytes and Mlkl knockout C57BL/6 mice. We demonstrated that arsenic exposure induces necroptosis by activating the receptor-interacting serine/threonine-protein kinase-3 (RIPK3)/MLKL pathway in vivo and in vitro. Consistent with our hypotheses, we found that necroptosis inhibitors (RIPK1 inhibitor necrostatin-1 [Nec-1], RIPK3 inhibitor [GSK-872], MLKL inhibitor [NSA]) and Mlkl genetic knockout can partially protect against arsenic-induced inflammatory damage. Additionally, these strategies can downregulate the expression of key proteins associated with the activation of the NLRP3 inflammasome, including NLRP3, Caspase-1, and interleukin-1β (IL-1β). Taken together, our findings demonstrate that MLKL triggers NLRP3 inflammasome activation and plays an essential role in arsenic-induced myocardial necroinflammation.
Collapse
Affiliation(s)
- Yixin Cui
- School of Public Health, Xi'an Jiaotong University Health Science Center, NHC Key Laboratory of Environment and Endemic Diseases, National Health Commission of the People's Republic of China, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, PR China.
| | - Meng Zhang
- School of Public Health, Xi'an Jiaotong University Health Science Center, NHC Key Laboratory of Environment and Endemic Diseases, National Health Commission of the People's Republic of China, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, PR China.
| | - Yi Xiao
- Shaanxi Provincial Center for Disease Control and Prevention, Xi'an 710054, PR China.
| | - Jinren Liu
- School of Public Health, Xi'an Jiaotong University Health Science Center, NHC Key Laboratory of Environment and Endemic Diseases, National Health Commission of the People's Republic of China, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, PR China.
| | - Yonghui Chen
- School of Public Health, Xi'an Jiaotong University Health Science Center, NHC Key Laboratory of Environment and Endemic Diseases, National Health Commission of the People's Republic of China, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, PR China.
| | - Xingran Ruan
- School of Public Health, Xi'an Jiaotong University Health Science Center, NHC Key Laboratory of Environment and Endemic Diseases, National Health Commission of the People's Republic of China, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, PR China.
| | - Xu Zhao
- School of Public Health, Xi'an Jiaotong University Health Science Center, NHC Key Laboratory of Environment and Endemic Diseases, National Health Commission of the People's Republic of China, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, PR China.
| | - Yinan Liu
- School of Public Health, Xi'an Jiaotong University Health Science Center, NHC Key Laboratory of Environment and Endemic Diseases, National Health Commission of the People's Republic of China, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, PR China.
| | - Yawen Shi
- School of Public Health, Xi'an Jiaotong University Health Science Center, NHC Key Laboratory of Environment and Endemic Diseases, National Health Commission of the People's Republic of China, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, PR China.
| | - Jing Tian
- School of Public Health, Xi'an Jiaotong University Health Science Center, NHC Key Laboratory of Environment and Endemic Diseases, National Health Commission of the People's Republic of China, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, PR China.
| | - Lian Li
- School of Public Health, Xi'an Jiaotong University Health Science Center, NHC Key Laboratory of Environment and Endemic Diseases, National Health Commission of the People's Republic of China, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, PR China.
| | - Xinhe Zhang
- School of Public Health, Xi'an Jiaotong University Health Science Center, NHC Key Laboratory of Environment and Endemic Diseases, National Health Commission of the People's Republic of China, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, PR China.
| | - Mingzhao Jia
- School of Public Health, Xi'an Jiaotong University Health Science Center, NHC Key Laboratory of Environment and Endemic Diseases, National Health Commission of the People's Republic of China, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, PR China.
| | - Yi Wang
- School of Public Health, Xi'an Jiaotong University Health Science Center, NHC Key Laboratory of Environment and Endemic Diseases, National Health Commission of the People's Republic of China, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, PR China.
| | - Xuewei Yang
- School of Public Health, Xi'an Jiaotong University Health Science Center, NHC Key Laboratory of Environment and Endemic Diseases, National Health Commission of the People's Republic of China, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, PR China.
| | - Zhaoxing Lin
- Shaanxi Provincial Center for Disease Control and Prevention, Xi'an 710054, PR China.
| | - Jinghong Chen
- School of Public Health, Xi'an Jiaotong University Health Science Center, NHC Key Laboratory of Environment and Endemic Diseases, National Health Commission of the People's Republic of China, No. 76 Yanta West Road, Xi'an, Shaanxi 710061, PR China.
| |
Collapse
|
4
|
Bi T, Koch H, Kremer B, Höllig A. Iron Hemostasis in Patients With Subarachnoid Hemorrhage and the Role of Early CSF Drainage. Neurology 2025; 105:e213767. [PMID: 40489716 DOI: 10.1212/wnl.0000000000213767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 04/03/2025] [Indexed: 06/11/2025] Open
Abstract
Early brain injury and delayed cerebral ischemia are combined intricate processes, and they represent the principal cause of subarachnoid hemorrhage (SAH)-related morbidity and mortality worldwide. Recent studies have shown that early lumbar CSF drainage can be used to decrease the incidence of delayed cerebral ischemia and improve long-term outcome. This approach has provided novel insights into post-SAH management that lessened the burden of secondary infarction and decreased the rate of unfavorable outcome. Given that the evaluation of this approach is contingent on prospective trial and early-stage randomized clinical trial, we review insights from studies that have elucidated the mechanisms underlying deterioration in SAH. We explore the role of iron homeostasis in the restoration of normal CSF circulation and the stabilization of optimal cerebral physiology to alleviate early brain injury and delayed neurologic impairment after SAH to advance the current understanding of SAH management.
Collapse
Affiliation(s)
- Tianshu Bi
- Department of Neurosurgery, Medical Faculty, RWTH Aachen University, Germany; and
| | - Henner Koch
- Department of Epileptology, Neurology, Medical Faculty, RWTH Aachen University, Germany
| | - Benedikt Kremer
- Department of Neurosurgery, Medical Faculty, RWTH Aachen University, Germany; and
| | - Anke Höllig
- Department of Neurosurgery, Medical Faculty, RWTH Aachen University, Germany; and
| |
Collapse
|
5
|
Han Y, Sun Y, Peng S, Tang T, Zhang B, Yu R, Sun X, Guo S, Ma L, Li P, Yang P. PI3K/AKT pathway: A potential therapeutic target in cerebral ischemia-reperfusion injury. Eur J Pharmacol 2025; 998:177505. [PMID: 40118329 DOI: 10.1016/j.ejphar.2025.177505] [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: 10/02/2024] [Revised: 02/21/2025] [Accepted: 03/10/2025] [Indexed: 03/23/2025]
Abstract
Cerebral ischemia is a prevalent cerebrovascular disorder, with the restoration of blocked blood vessels serving as the current standard clinical treatment. However, reperfusion can exacerbate neuronal damage and neurological dysfunction, resulting in cerebral ischemia-reperfusion (I/R) injury. Presently, clinical treatment strategies for cerebral I/R injury are limited, creating an urgent need to identify new effective therapeutic targets. The PI3K/AKT signaling pathway, a pro-survival pathway associated with cerebral I/R injury, has garnered significant attention. We conducted a comprehensive review of the literature on the PI3K/AKT pathway in the context of cerebral I/R. Our findings indicate that activation of the PI3K/AKT signaling pathway following cerebral I/R can alleviate oxidative stress, reduce endoplasmic reticulum stress (ERS), inhibit inflammatory responses, decrease neuronal apoptosis, autophagy, and pyroptosis, mitigate blood-brain barrier (BBB) damage, and promote neurological function recovery. Consequently, this pathway ultimately reduces neuronal death, alleviates brain tissue damage, decreases the volume of cerebral infarction, and improves behavioral impairments. These results suggest that the PI3K/AKT signaling pathway is a promising therapeutic target for further research and drug development, holding significant potential for the treatment of cerebral I/R injury.
Collapse
Affiliation(s)
- Yiming Han
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Yu Sun
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Shiyu Peng
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Tingting Tang
- First Clinical College, Xinxiang Medical University, Xinxiang, China
| | - Beibei Zhang
- First Clinical College, Xinxiang Medical University, Xinxiang, China
| | - Ruonan Yu
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Xiaoyan Sun
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China
| | - Shanshan Guo
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China; Staff Hospital of Henan Fifth Construction Group Co., Ltd, Zhengzhou, Henan, China
| | - Lijuan Ma
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China.
| | - Peng Li
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China.
| | - Pengfei Yang
- College of Pharmacy, Xinxiang Medical University, Henan international Joint Laboratory of Cardiovascular Remodeling and Drug intervention, China; Xinxiang Key Laboratory of Vascular Remodeling intervention and Molecular Targeted Therapy Drug Development, Xinxiang, China.
| |
Collapse
|
6
|
Gong HB, Yan CY, Ye HE, Tan SY, Chen SS, Ye JW, Sun WY, Tan HL, Luo X, Niu J, Li K, Wang JH, Kurihara H, Li YF, He RR. Screening novel anti-inflammatory peptides inhibiting the NLRP3 inflammasome from UHPLC-MS/MS-characterized peptide profiles of cocoa tea protein hydrolysates. Food Res Int 2025; 212:116519. [PMID: 40382080 DOI: 10.1016/j.foodres.2025.116519] [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: 02/05/2025] [Revised: 03/31/2025] [Accepted: 04/21/2025] [Indexed: 05/20/2025]
Abstract
Cocoa tea (Camellia ptilophylla) is a popular beverage enjoyed worldwide, yet its protein-rich residues are often underutilized during processing. In this study, proteins from cocoa tea were extracted and hydrolyzed with alkaline protease to produce cocoa tea protein hydrolysates (CTPH). The results showed that CTPH exhibited promising anti-inflammatory activity. To uncover bioactive peptides, the hydrolysates were analyzed using UHPLC-MS/MS, and potential peptides were screened through molecular docking targeting the NLRP3 inflammasome. In an in vitro model of NLRP3 inflammasome activation, two active peptides, LLR and LIGF, were found to significantly reduce IL-1β production in PMA-differentiated THP-1 macrophages following treatment with nigericin or ATP. These peptides interact with the NACHT domain of NLRP3 via hydrogen bonding and hydrophobic interactions. Their binding to NLRP3 was further confirmed by CETSA assay. These findings suggest that cocoa tea-derived peptides could offer therapeutic potential for managing inflammation.
Collapse
Affiliation(s)
- Hai-Biao Gong
- Department of Pharmacy, The First Affiliated Hospital, Jinan University, Guangzhou 510632, China / College of Pharmacy, Jinan University, Guangzhou 510632, China; Guangzhou Key Laboratory of State Key Laboratory of Bioactive Molecules and Druggability Assessment/Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs/Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility/Guangdong Engineering Research Center of Traditional Chinese Medicine & Health Products/International Cooperative Laboratory of TCM Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/The Second Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510632, China
| | - Chang-Yu Yan
- Department of Pharmacy, The First Affiliated Hospital, Jinan University, Guangzhou 510632, China / College of Pharmacy, Jinan University, Guangzhou 510632, China; Guangzhou Key Laboratory of State Key Laboratory of Bioactive Molecules and Druggability Assessment/Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs/Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility/Guangdong Engineering Research Center of Traditional Chinese Medicine & Health Products/International Cooperative Laboratory of TCM Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/The Second Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510632, China
| | - Hui-Er Ye
- Department of Pharmacy, The First Affiliated Hospital, Jinan University, Guangzhou 510632, China / College of Pharmacy, Jinan University, Guangzhou 510632, China; Guangzhou Key Laboratory of State Key Laboratory of Bioactive Molecules and Druggability Assessment/Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs/Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility/Guangdong Engineering Research Center of Traditional Chinese Medicine & Health Products/International Cooperative Laboratory of TCM Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/The Second Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510632, China
| | - Shuo-Yan Tan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314100, China
| | - Shi-Sheng Chen
- Guangzhou Springsnow Biotechnology Co., Ltd, Guangzhou 510670, China
| | - Jian-Wen Ye
- Guangzhou Springsnow Biotechnology Co., Ltd, Guangzhou 510670, China
| | - Wan-Yang Sun
- Department of Pharmacy, The First Affiliated Hospital, Jinan University, Guangzhou 510632, China / College of Pharmacy, Jinan University, Guangzhou 510632, China; Guangzhou Key Laboratory of State Key Laboratory of Bioactive Molecules and Druggability Assessment/Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs/Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility/Guangdong Engineering Research Center of Traditional Chinese Medicine & Health Products/International Cooperative Laboratory of TCM Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/The Second Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510632, China
| | - Hong-Li Tan
- Guangzhou Key Laboratory of State Key Laboratory of Bioactive Molecules and Druggability Assessment/Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs/Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility/Guangdong Engineering Research Center of Traditional Chinese Medicine & Health Products/International Cooperative Laboratory of TCM Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/The Second Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510632, China
| | - Xiang Luo
- Guangzhou Key Laboratory of State Key Laboratory of Bioactive Molecules and Druggability Assessment/Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs/Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility/Guangdong Engineering Research Center of Traditional Chinese Medicine & Health Products/International Cooperative Laboratory of TCM Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/The Second Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510632, China; Department of Food Science and Engineering, Jinan University, Guangzhou 510632, Guangdong, China
| | - Jie Niu
- Guangzhou Key Laboratory of State Key Laboratory of Bioactive Molecules and Druggability Assessment/Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs/Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility/Guangdong Engineering Research Center of Traditional Chinese Medicine & Health Products/International Cooperative Laboratory of TCM Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/The Second Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510632, China
| | - Kun Li
- Guangzhou Key Laboratory of State Key Laboratory of Bioactive Molecules and Druggability Assessment/Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs/Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility/Guangdong Engineering Research Center of Traditional Chinese Medicine & Health Products/International Cooperative Laboratory of TCM Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/The Second Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510632, China
| | - Jing-Hao Wang
- Department of Pharmacy, The First Affiliated Hospital, Jinan University, Guangzhou 510632, China / College of Pharmacy, Jinan University, Guangzhou 510632, China.
| | - Hiroshi Kurihara
- Department of Pharmacy, The First Affiliated Hospital, Jinan University, Guangzhou 510632, China / College of Pharmacy, Jinan University, Guangzhou 510632, China; Guangzhou Key Laboratory of State Key Laboratory of Bioactive Molecules and Druggability Assessment/Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs/Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility/Guangdong Engineering Research Center of Traditional Chinese Medicine & Health Products/International Cooperative Laboratory of TCM Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/The Second Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510632, China.
| | - Yi-Fang Li
- Department of Pharmacy, The First Affiliated Hospital, Jinan University, Guangzhou 510632, China / College of Pharmacy, Jinan University, Guangzhou 510632, China; Guangzhou Key Laboratory of State Key Laboratory of Bioactive Molecules and Druggability Assessment/Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs/Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility/Guangdong Engineering Research Center of Traditional Chinese Medicine & Health Products/International Cooperative Laboratory of TCM Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/The Second Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510632, China.
| | - Rong-Rong He
- Department of Pharmacy, The First Affiliated Hospital, Jinan University, Guangzhou 510632, China / College of Pharmacy, Jinan University, Guangzhou 510632, China; Guangzhou Key Laboratory of State Key Laboratory of Bioactive Molecules and Druggability Assessment/Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs/Guangdong Engineering Research Center of Traditional Chinese Medicine & Disease Susceptibility/Guangdong Engineering Research Center of Traditional Chinese Medicine & Health Products/International Cooperative Laboratory of TCM Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/The Second Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510632, China; State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China.
| |
Collapse
|
7
|
Zhong S, Shen H, Dai X, Liao L, Huang C. BAM15 inhibits endothelial pyroptosis via the NLRP3/ASC/caspase-1 pathway to alleviate atherosclerosis. Atherosclerosis 2025; 406:119226. [PMID: 40393254 DOI: 10.1016/j.atherosclerosis.2025.119226] [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: 11/07/2024] [Revised: 04/08/2025] [Accepted: 04/23/2025] [Indexed: 05/22/2025]
Abstract
BACKGROUND AND AIMS Atherosclerosis (AS) is a chronic inflammatory disease contributing to major cardiovascular events. This study aimed to investigate the effects of BAM15, a mitochondrial uncoupler, on regulating the NLRP3/ASC/caspase-1 signaling pathway to suppress endothelial cell pyroptosis and mitigate AS. METHODS AS was induced in ApoE-/- mice through a high-fat diet (HFD), and the therapeutic effects of BAM15 (5 mg/kg/day, s. c.) were evaluated. Histological analyses, including HE staining and oil red O staining, were used to assess aortic pathology and lipid deposition. Serum inflammatory cytokines (IL-1β, IL-18) were quantified by ELISA. Mouse primary aortic endothelial cells (MAECs) were treated with oxidized low-density lipoprotein (ox-LDL) to simulate AS condition in vitro. Mitochondrial reactive oxygen species (mtROS) expression and oxidized (ox)-mtDNA content were detected by Mitosox staining and ELISA, respectively. Western blot was used to assess the expression of pyroptosis-related proteins, including GSDMD-NT, NLRP3, ASC, and cleaved-caspase-1. RESULTS BAM15 reduced atherosclerotic plaque formation, lipid deposition, and inflammation, and diminished mtROS expression and ox-mtDNA content in the AS mouse models. In both in vivo and in vitro experiments, BAM15 markedly inhibited the activation of the NLRP3 inflammasome, leading to reduced pyroptosis in endothelial cells. Activation of the NLRP3/ASC/caspase-1 signaling pathway by Nigericin partially reversed the protective effects of BAM15, underscoring the pivotal role of NLRP3 inflammasome inhibition in endothelial pyroptosis suppression. CONCLUSIONS BAM15 effectively inhibits endothelial cell pyroptosis by reducing mtROS production and ox-mtDNA release to suppress the NLRP3/ASC/caspase-1 signaling pathway, thereby alleviating AS in both in vivo and in vitro models.
Collapse
Affiliation(s)
- Su Zhong
- Department of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China; Cadre Ward, Army 73rd Group Military Hospital, Xiamen, 36100, Fujian, China
| | - Hui Shen
- Department of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
| | - Xiaoman Dai
- Department of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
| | - Lianming Liao
- Department of Laboratory Medicine, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
| | - Chun Huang
- Department of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China; People's Hospital of Changji Hui Autonomous Prefecture, Changji, 831100, Xinjiang, China.
| |
Collapse
|
8
|
Yang L, Mo W, Xin L, Zhang M, Chen K, Guo X, Zhang J, Yu B. Rescuing fertility: Itaconic acid prevents ovarian damage through NRF2-mediated pyroptosis pathways in diminished ovarian reserve models. Cell Signal 2025; 131:111766. [PMID: 40147551 DOI: 10.1016/j.cellsig.2025.111766] [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: 12/20/2024] [Revised: 03/06/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Diminished ovarian reserve (DOR) is a major cause of infertility, often triggered by inflammation and oxidative stress. Pyroptosis, a form of programmed cell death, has been implicated in DOR pathogenesis. Itaconic acid (IA), an endogenous metabolite, is known for its anti-inflammatory and antioxidant properties. This study aimed to explore whether IA could alleviate lipopolysaccharide (LPS)-induced DOR in mice by inhibiting pyroptosis through the NRF2 pathway. METHODS A DOR mouse model was established by administering LPS for 5 consecutive days, followed by IA treatment. Ovarian function was assessed by follicle count and hormone levels. Inflammatory markers, oxidative stress, and pyroptosis-related proteins were evaluated in both in vivo and in vitro models. The molecular mechanism was further investigated using inhibitors and molecular docking studies. RESULTS IA significantly improved ovarian function in LPS-induced DOR mice by increasing the number of follicles and normalizing hormone levels. IA also reduced inflammation, oxidative stress, and pyroptosis, as evidenced by lower expression of NLRP3, cleaved-caspase-1, and N-GSDMD, while increasing NRF2 expression. In vitro, IA enhanced granulosa cell (GC) viability, reduced reactive oxygen species (ROS), and decreased pyroptosis in LPS-treated GCs. Additionally, the beneficial effects of IA were mediated via the NRF2 pathway, as NRF2 inhibition (ML385) reversed these improvements. Additionally, we identified GSDMD as a downstream target of IA, with inhibition of GSDMD ameliorating DOR progression and inflammatory responses. CONCLUSION IA alleviates LPS-induced DOR by reducing inflammation, oxidative stress, and pyroptosis through activation of the NRF2 signaling and direct inhibition of the GSDMD pathway. These findings suggest that IA may serve as a potential therapeutic agent for improving ovarian reserve and fertility.
Collapse
Affiliation(s)
- Ling Yang
- Department of Obstetrics and Gynecology, the Hexian People's Hospital, Maanshan 238200, Anhui, China
| | - Wenya Mo
- School of Nursing, Anhui Medical University, Hefei 230032, Anhui, China; Department of Urology, the First Affiliated Hospital of University of Science and Technology of China, Hefei 230036, Anhui, China
| | - Lei Xin
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei 230032, Anhui, China; Engineering Research Center of Biopreservation and Artificial Organs, Ministry of Education, Hefei 230032, Anhui, China
| | - Mingzhao Zhang
- Department of Breast Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui, China
| | - Kegong Chen
- Department of Thoracic Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui, China
| | - Xiaohui Guo
- Department of Pathology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui, China.
| | - Jing Zhang
- College of Veterinary Medicine, Jilin University, Changchun 130062, Jilin, China.
| | - Biao Yu
- Department of Obstetrics and Gynecology, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Medical University, Hefei 230032, Anhui, China; Engineering Research Center of Biopreservation and Artificial Organs, Ministry of Education, Hefei 230032, Anhui, China.
| |
Collapse
|
9
|
Duan WL, Gu LH, Guo A, Wang XJ, Ding YY, Zhang P, Zhang BG, Li Q, Yang LX. Molecular mechanisms of programmed cell death and potential targeted pharmacotherapy in ischemic stroke (Review). Int J Mol Med 2025; 56:103. [PMID: 40341937 PMCID: PMC12081036 DOI: 10.3892/ijmm.2025.5544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 04/15/2025] [Indexed: 05/11/2025] Open
Abstract
Stroke poses a threat to the elderly, being the second leading cause of death and the third leading cause of disability worldwide. Ischemic stroke (IS), resulting from arterial occlusion, accounts for ~85% of all strokes. The pathophysiological processes involved in IS are intricate and complex. Currently, tissue plasminogen activator (tPA) is the only Food and Drug Administration‑approved drug for the treatment of IS. However, due to its limited administration window and the risk of symptomatic hemorrhage, tPA is applicable to only ~10% of patients with stroke. Additionally, the reperfusion process associated with thrombolytic therapy can further exacerbate damage to brain tissue. Therefore, a thorough understanding of the molecular mechanisms underlying IS‑induced injury and the identification of potential protective agents is critical for effective IS treatment. Over the past few decades, advances have been made in exploring potential protective drugs for IS. The present review summarizes the specific mechanisms of various forms of programmed cell death (PCD) induced by IS and highlights potential protective drugs targeting different PCD pathways investigated over the last decade. The present review provides a theoretical foundation for basic research and insights for the development of pharmacotherapy for IS.
Collapse
Affiliation(s)
- Wan-Li Duan
- Medical Research Center, Shaoxing People's Hospital, Shaoxing, Zhejiang 312000, P.R. China
- Department of Diagnostic Pathology, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong 261041, P.R. China
| | - Li-Hui Gu
- Department of Diagnostic Pathology, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong 261041, P.R. China
| | - Ai Guo
- Department of Diagnostic Pathology, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong 261041, P.R. China
| | - Xue-Jie Wang
- Department of Diagnostic Pathology, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong 261041, P.R. China
- Department of Pathology, Shaoxing People's Hospital, Shaoxing, Zhejiang 312000, P.R. China
| | - Yi-Yue Ding
- Department of Diagnostic Pathology, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong 261041, P.R. China
| | - Peng Zhang
- Department of Cardiology, Shaoxing People's Hospital, Shaoxing, Zhejiang 312000, P.R. China
| | - Bao-Gang Zhang
- Department of Diagnostic Pathology, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, Shandong 261041, P.R. China
- Department of Pathology, Shaoxing People's Hospital, Shaoxing, Zhejiang 312000, P.R. China
| | - Qin Li
- Rehabilitation Medicine and Health College, Hunan University of Medicine, Huaihua, Hunan 418000, P.R. China
| | - Li-Xia Yang
- Medical Research Center, Shaoxing People's Hospital, Shaoxing, Zhejiang 312000, P.R. China
| |
Collapse
|
10
|
Chen X, Zheng Y, Zhang X, Zheng A, Huang J, Deng G, Wu X, Peng Y, Zhang X, Chen R, Xiao Q, Ye W. Harnessing FDA-approved dipyridamole to inhibit NLRP3 inflammasome and improve outcomes of acute lung injury in sepsis. Toxicol Appl Pharmacol 2025; 500:117383. [PMID: 40360054 DOI: 10.1016/j.taap.2025.117383] [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: 11/19/2024] [Revised: 05/01/2025] [Accepted: 05/06/2025] [Indexed: 05/15/2025]
Abstract
Aberrant activation of the NLRP3 inflammasome is critically involved in sepsis-induced acute lung injury (ALI), with inhibition of this pathway emerging as a promising therapeutic approach. This study identifies Dipyridamole, an FDA-approved drug, as a novel inhibitor of the NLRP3 inflammasome. Mechanistically, Dipyridamole suppresses mitochondrial ROS release and directly interacts with NEK7, thereby preventing its association with NLRP3 and impeding inflammasome complex assembly. In an LPS-induced sepsis model, Dipyridamole significantly ameliorated ALI, reduced inflammatory responses, and improved survival rates in model mice. Additionally, Dipyridamole effectively inhibited NLRP3 inflammasome activation in lung tissue. These findings position Dipyridamole as a potent NLRP3 inflammasome inhibitor with substantial therapeutic potential for managing sepsis-induced ALI.
Collapse
Affiliation(s)
- Xiuhui Chen
- Department of Pharmacy & the Eighth People's Hospital of Dongguan, Dongguan Children's Hospital Affiliated to Guangdong Medical University, Dongguan 523000, China; Key Laboratory of Precision Pharmacy and Pharmaceutical Basic Research, Dongguan Institute of Pediatrics, Dongguan 523000, China
| | - Yutong Zheng
- Department of Pharmacy & the Eighth People's Hospital of Dongguan, Dongguan Children's Hospital Affiliated to Guangdong Medical University, Dongguan 523000, China; Key Laboratory of Precision Pharmacy and Pharmaceutical Basic Research, Dongguan Institute of Pediatrics, Dongguan 523000, China
| | - Xiaofeng Zhang
- Department of Pharmacy & the Eighth People's Hospital of Dongguan, Dongguan Children's Hospital Affiliated to Guangdong Medical University, Dongguan 523000, China; Key Laboratory of Precision Pharmacy and Pharmaceutical Basic Research, Dongguan Institute of Pediatrics, Dongguan 523000, China
| | - Anran Zheng
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou 511436, China
| | - Junjun Huang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou 511436, China
| | - Guoliang Deng
- Department of Pharmacy & the Eighth People's Hospital of Dongguan, Dongguan Children's Hospital Affiliated to Guangdong Medical University, Dongguan 523000, China; Key Laboratory of Precision Pharmacy and Pharmaceutical Basic Research, Dongguan Institute of Pediatrics, Dongguan 523000, China
| | - Xuna Wu
- Department of Pharmacy & the Eighth People's Hospital of Dongguan, Dongguan Children's Hospital Affiliated to Guangdong Medical University, Dongguan 523000, China; Key Laboratory of Precision Pharmacy and Pharmaceutical Basic Research, Dongguan Institute of Pediatrics, Dongguan 523000, China
| | - Yuying Peng
- Department of Pharmacy & the Eighth People's Hospital of Dongguan, Dongguan Children's Hospital Affiliated to Guangdong Medical University, Dongguan 523000, China
| | - Xiaoling Zhang
- Maternal and Children Hospital of Guangdong Province, Guangzhou 511436, China
| | - Renshan Chen
- Guangzhou Hospital of Integrated Traditional and Western Medicine, Guangzhou 511436, China.
| | - Qing Xiao
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou 511436, China.
| | - Weijun Ye
- Department of Pharmacy & the Eighth People's Hospital of Dongguan, Dongguan Children's Hospital Affiliated to Guangdong Medical University, Dongguan 523000, China; Key Laboratory of Precision Pharmacy and Pharmaceutical Basic Research, Dongguan Institute of Pediatrics, Dongguan 523000, China.
| |
Collapse
|
11
|
Liu H, Xue H, Guo Q, Xue X, Yang L, Zhao K, Liu Y. Ferroptosis meets inflammation: A new frontier in cancer therapy. Cancer Lett 2025; 620:217696. [PMID: 40189012 DOI: 10.1016/j.canlet.2025.217696] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 03/26/2025] [Accepted: 04/03/2025] [Indexed: 04/10/2025]
Abstract
Ferroptosis, an iron-dependent form of regulated cell death driven by lipid peroxidation, has emerged as a critical player in cancer pathogenesis. Concurrently, inflammation, a key biological response to tissue injury or infection, significantly influences cancer development and progression. The interplay between ferroptosis and inflammation represents a promising yet underexplored area of research. This review synthesizes recent advances in understanding the molecular mechanisms governing their interaction, emphasizing how ferroptosis triggers inflammatory responses and how inflammatory mediators, such as TNF-α, regulate ferroptosis through iron metabolism and lipid peroxidation pathways. Key molecular targets within the ferroptosis-inflammation axis, including GPX4, ACSL4, and the NF-κB signaling pathway, offer therapeutic potential for cancer treatment. By modulating these targets, it may be possible to enhance ferroptosis and fine-tune inflammatory responses, thereby improving therapeutic outcomes. Additionally, this review explores the broader implications of targeting the ferroptosis-inflammation interplay in disease treatment, highlighting opportunities for developing innovative strategies to combat cancer. By bridging the gap in current knowledge, this review provides a comprehensive resource for researchers and clinicians, offering insights into the therapeutic potential of this intricate biological relationship.
Collapse
Affiliation(s)
- Hu Liu
- Department of Oncology Surgery, Shanghai Mengchao Hospital, Shanghai University, Shanghai, 202800, China
| | - Hui Xue
- Department of Oncology Surgery, Shanghai Mengchao Hospital, Shanghai University, Shanghai, 202800, China
| | - Qian Guo
- Department of Rhinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Xutong Xue
- Boston Children's Hospital, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Lixue Yang
- Department of Oncology Surgery, Shanghai Mengchao Hospital, Shanghai University, Shanghai, 202800, China.
| | - Kaijun Zhao
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| | - Yu'e Liu
- Boston Children's Hospital, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA; Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| |
Collapse
|
12
|
Li Y, Zhou B, Liu D, Nie G, Yang F, Chen J, Cheng S, Kang Y, Liu B, Dong B, Liu M. Carbon monoxide gas molecules: Therapeutic mechanisms in radiation-induced lung injury. J Colloid Interface Sci 2025; 688:250-263. [PMID: 40010090 DOI: 10.1016/j.jcis.2025.02.126] [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: 12/15/2024] [Revised: 02/17/2025] [Accepted: 02/18/2025] [Indexed: 02/28/2025]
Abstract
Radiation therapy (RT) remains an essential treatment modality for lung cancer, yet its effectiveness is frequently hindered by radiation-induced lung injury (RILI), a common outcome of modern therapeutic regimens. With the aim of addressing this challenge, a novel nanocomposite, Au@mSiO2@Mn(CO)5Br (ASMB), was synthesized with Au@mSiO2 as the carrier and Mn(CO)5Br as the functional component. The gold nanorods (Au rods) core generates reactive oxygen species (ROS) under X-ray irradiation, which then activates Mn(CO)5Br to release carbon monoxide (CO) locally within the lung during radiotherapy. The released CO then diffuses to surrounding tissues, inhibiting the excessive accumulation of ROS, thereby preventing damage to normal cells caused by ROS generated in a short period of time. Meanwhile, the released manganese ions (Mnn+) catalyze the conversion of hydrogen peroxide (H2O2) in the microenvironment into oxygen (O2). In vitro experiments demonstrated that the release of CO markedly attenuated radiation-induced ROS production, thereby inhibiting the activation of the NLRP3 inflammasome and reducing the levels of inflammatory cytokines and pyroptosis-related proteins. Moreover, it downregulated the expression of fibrosis-associated proteins, including TGF-β1 and α-SMA. Additionally, CO facilitated DNA damage repair, thereby mitigating radiation-induced tissue injury. In the RILI model, the ASMB NPs-treated lungs exhibited notably reduced pulmonary edema, congestion, and inflammatory cell infiltration, primarily by inhibiting NLRP3 inflammasome-dependent pyroptosis and reducing levels of inflammation and fibrosis markers. The release of O2 further mitigates local tissue hypoxia, enhancing the effectiveness of radiotherapy. Overall, ASMB NPs provide a promising alternative for the treatment of RILI and a potential therapeutic strategy to improve the efficacy of radiotherapy.
Collapse
Affiliation(s)
- Ya'nan Li
- Department of Radiation Oncology, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei, Anhui 230601, China
| | - Bingshuai Zhou
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, Changchun 130021, China
| | - Dajie Liu
- Department of Radiation Oncology, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei, Anhui 230601, China
| | - Guodong Nie
- Department of Radiation Oncology, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei, Anhui 230601, China
| | - Fan Yang
- Department of Radiation Oncology, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei, Anhui 230601, China
| | - Jiajie Chen
- Department of Radiation Oncology, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei, Anhui 230601, China
| | - Sen Cheng
- Department of Radiation Oncology, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei, Anhui 230601, China
| | - Yahui Kang
- Department of Radiation Oncology, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei, Anhui 230601, China; Department of Radiation Oncology, The First Affiliated Hospital of University of Science and Technology of China, Hefei, Anhui 230031, China
| | - Bailong Liu
- Department of Radiation Oncology, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei, Anhui 230601, China.
| | - Biao Dong
- State Key Laboratory on Integrated Optoelectronics, College of Electronic Science and Engineering, Jilin University, Changchun 130021, China.
| | - Min Liu
- Department of Radiation Oncology, The Second Affiliated Hospital of Anhui Medical University, 678 Furong Road, Hefei, Anhui 230601, China.
| |
Collapse
|
13
|
Li W, Li Y, Qiu Y, Huang R, Niu J, Chen J, Liu Y, Chen L. Kurarinone and Nor-kurarinone inhibit NLRP3 inflammasome activation and regulate macrophage polarization against ulcerative colitis. Int Immunopharmacol 2025; 157:114758. [PMID: 40318276 DOI: 10.1016/j.intimp.2025.114758] [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: 10/16/2024] [Revised: 04/01/2025] [Accepted: 04/26/2025] [Indexed: 05/07/2025]
Abstract
Activation of NOD-like receptor protein 3 (NLRP3) can lead to the production of inflammatory factors and perturbation of macrophage polarization, leading to an intestinal immune imbalance that promotes the progression of ulcerative colitis (UC). In this study, we investigated the therapeutic effect of Kurarinone and Nor-kurarinone on UC and their regulatory mechanisms relating to NLRP3 inflammasome activation and macrophage polarization. UC mice were induced using dextran sulfate sodium (DSS) and treated with Kurarinone and Nor-kurarinone. Results showed that Kurarinone and Nor-kurarinone could alleviate weight loss, decrease the disease activity index (DAI) score, shorten colon length, inhibit formation of the NLRP3 inflammasome in the colon and regulate macrophage polarization in UC mice. The THP-1 cells were used as an in vitro model of the NLRP3 inflammasome, conducted by treatment with lipopolysaccharide (LPS) and ATP/Nigericin. Kurarinone and Nor-kurarinone can inhibit the NLRP3 inflammasome formation response by disrupting the NLRP3/ASC interaction to inhibit NLRP3 assembly and then regulating the polarization of macrophages. In conclusion, Kurarinone and Nor-kurarinone inhibited NLRP3 inflammasome assembly to counteract activation of the NLRP3 inflammasome. This inhibition led to a reduction in M1 polarization of intestinal macrophages in UC mice to keep the balance of M1/ M2 macrophages. Our study suggests that Kurarinone and Nor-kurarinone may be novel therapeutic modalities for UC.
Collapse
Affiliation(s)
- Wanyu Li
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yadi Li
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yongyi Qiu
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Ruiting Huang
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jing Niu
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jiawen Chen
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yi Liu
- School of Chinese Medicine, Southern Medical University, Guangzhou 510515, China.
| | - Lei Chen
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
14
|
Qiu M, Hao Z, Liu Y, Liu Y, Chang M, Lin X, Liu X, Dong N, Sun W, Teng X. ROS acted as an initial role in selenium nanoparticles alleviating insecticide chlorpyrifos-induced oxidative stress, pyroptosis, and intestinal barrier dysfunction in porcine intestinal epithelial cells. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2025; 211:106418. [PMID: 40350231 DOI: 10.1016/j.pestbp.2025.106418] [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: 11/17/2024] [Revised: 02/03/2025] [Accepted: 04/14/2025] [Indexed: 05/14/2025]
Abstract
Chlorpyrifos (CPF), a toxic organophosphorus insecticide, is widely used in agriculture to protect crops (eg., maize) from pests. The use of CPF in crops can result in accumulation in crop seeds, such as corn seeds, which is a primary feed ingredient in pigs. Pigs in China, which is an important source of animal-derived protein in the Chinese diet, account for over 50 % of the raised pig population in the whole world. Therefore, CPF may pose a potential risk to the health of non-target organisms (pigs and humans) through the food chain. However, whether CPF can damage porcine intestine remains unknown. Selenium (Se), an essential trace element, was reported to have antioxidant and anti-toxic effects. Tight junction (TJ) is an important mechanism of intestinal injury and pyroptosis is a new hotspot in the field of toxicology. Hence, we wanted to investigate whether CPF can damage pig intestine and whether selenium nanoparticles (SeNPs) supplement can alleviate CPF-induced pig intestine damage, and to study corresponding mechanism from the three aspects of OS, pytoptosis, and TJ. We established a model of SeNPs alleviating damage caused by CPF in intestinal porcine enterocytes (IPEC-J2 cells), and found that SeNPs alleviated CPF-induced oxidative stress (OS), pyroptosis, and intestinal barrier dysfunction in IPEC-J2 cells. Interestingly, OS, pyroptosis, and intestinal barrier dysfunction had serial relations, and ROS/Nrf2/Caspase-1/Occludin and ROS/Nrf2/Caspase-1/ZO-1 pathways played a role. Notably, ROS and Caspase-1 played an initial and important role, respectively. Our study added new information on pesticides-caused damage to non-target organisms, and provided new idea, insight, and targets to mitigatie pesticides-induced toxic effect on non-target organisms.
Collapse
Affiliation(s)
- Minna Qiu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China
| | - Zhiyu Hao
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China
| | - Yuhao Liu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China
| | - Yuhang Liu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China
| | - Minghang Chang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China
| | - Xu Lin
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China
| | - Xiumei Liu
- College of Life Sciences, Yantai University, Yantai 264005, PR China
| | - Na Dong
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China.
| | - Wei Sun
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China.
| | - Xiaohua Teng
- College of Animal Science and Technology, Northeast Agricultural University, Harbin 150030, PR China.
| |
Collapse
|
15
|
Ayyubova G, Madhu LN. Microglial NLRP3 Inflammasomes in Alzheimer's Disease Pathogenesis: From Interaction with Autophagy/Mitophagy to Therapeutics. Mol Neurobiol 2025; 62:7124-7143. [PMID: 39951189 DOI: 10.1007/s12035-025-04758-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 02/08/2025] [Indexed: 05/15/2025]
Abstract
The nucleotide-binding oligomerization domain-like receptor pyrin domain-containing 3 (NLRP3) inflammasome, discovered 20 years ago, is crucial in controlling innate immune reactions in Alzheimer's disease (AD). By initiating the release of inflammatory molecules (including caspases, IL-1β, and IL-18), the excessively activated inflammasome complex in microglia leads to chronic inflammation and neuronal death, resulting in the progression of cognitive deficiencies. Even though the involvement of NLRP3 has been implicated in neuroinflammation and widely explored in several studies, there are plenty of controversies regarding its precise roles and activation mechanisms in AD. Another prominent feature of AD is impairment in microglial autophagy, which can be either the cause or the consequence of NLRP3 activation and contributes to the aggregation of misfolded proteins and aberrant chronic inflammatory state seen in the disease course. Studies also demonstrate that intracellular buildup of dysfunctional and damaged mitochondria due to defective mitophagy enhances inflammasome activation, further suggesting that restoration of impaired autophagy and mitophagy can effectively suppress it, thereby reducing inflammation and protecting microglia and neurons. This review is primarily focused on the role of NLRP3 inflammasome in the etiopathology of AD, its interactions with microglial autophagy/mitophagy, and the latest developments in NLRP3 inflammasome-targeted therapeutic interventions being implicated for AD treatment.
Collapse
Affiliation(s)
- Gunel Ayyubova
- Department of Cytology, Embryology and Histology, Azerbaijan Medical University, Baku, Azerbaijan.
| | - Leelavathi N Madhu
- Institute for Regenerative Medicine, Department of Cell Biology and Genetics, Texas A&M Health Science Center School of Medicine, College Station, TX, USA
| |
Collapse
|
16
|
Liu J, Li Z, Li Z, Wang A, Liao X, Liu Z, Wu J. Fudosteine attenuates lung inflammation in mice with PM2.5-induced asthma exacerbation by inhibiting pyroptosis via the NLRP3/caspase-1/GSDMD pathway. Toxicol Appl Pharmacol 2025; 499:117346. [PMID: 40228672 DOI: 10.1016/j.taap.2025.117346] [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: 04/09/2025] [Accepted: 04/11/2025] [Indexed: 04/16/2025]
Abstract
This study aimed to explore the potential preventive effects of fudosteine (Fud) on PM2.5-induced asthma exacerbations in a murine model. BALB/c mice were randomly allocated into six groups: control, Fud, ovalbumin (OVA), OVA+Fud, OVA+PM2.5, and OVA+PM2.5 + Fud. An asthma model was established through OVA sensitization and challenge. Compared to the OVA group, PM2.5 exposure exacerbated allergic asthma, as evidenced by increased collagen fiber deposition, goblet cell metaplasia, mucus secretion, heightened airway inflammation, elevated total cell and eosinophil counts, and upregulated levels of interleukin (IL)-1β, IL-18, and NLRP3 expression in lung tissues. Notably, fudosteine treatment mitigated these pathological changes. Western blot analysis revealed that fudosteine significantly reduced the expression of NLRP3, caspase-1, gasdermin D (GSDMD), cleaved-caspase-1, and cleaved-GSDMD in lung tissues. In conclusion, fudosteine alleviated lung inflammation, collagen deposition, and mucus secretion in PM2.5-induced asthma exacerbation, potentially by inhibiting the NLRP3 inflammasome-mediated pyroptosis pathway.
Collapse
Affiliation(s)
- Jianling Liu
- School of Medicine South China University of Technology, Guangzhou, Guangdong 510000, China; Second Department of Elderly Respiratory, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Zhongpeng Li
- Second Department of Elderly Respiratory, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China; Critical Care Medicine Department, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Zhangwen Li
- School of Medicine South China University of Technology, Guangzhou, Guangdong 510000, China; Second Department of Elderly Respiratory, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Aili Wang
- Second Department of Elderly Respiratory, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Xiaoyang Liao
- Second Department of Elderly Respiratory, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Zhangquan Liu
- Second Department of Elderly Respiratory, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Jian Wu
- School of Medicine South China University of Technology, Guangzhou, Guangdong 510000, China; Second Department of Elderly Respiratory, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China.
| |
Collapse
|
17
|
Paik S, Kim JK, Shin HJ, Park EJ, Kim IS, Jo EK. Updated insights into the molecular networks for NLRP3 inflammasome activation. Cell Mol Immunol 2025; 22:563-596. [PMID: 40307577 DOI: 10.1038/s41423-025-01284-9] [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: 12/24/2024] [Accepted: 03/17/2025] [Indexed: 05/02/2025] Open
Abstract
Over the past decade, significant advances have been made in our understanding of how NACHT-, leucine-rich-repeat-, and pyrin domain-containing protein 3 (NLRP3) inflammasomes are activated. These findings provide detailed insights into the transcriptional and posttranslational regulatory processes, the structural-functional relationship of the activation processes, and the spatiotemporal dynamics of NLRP3 activation. Notably, the multifaceted mechanisms underlying the licensing of NLRP3 inflammasome activation constitute a focal point of intense research. Extensive research has revealed the interactions of NLRP3 and its inflammasome components with partner molecules in terms of positive and negative regulation. In this Review, we provide the current understanding of the complex molecular networks that play pivotal roles in regulating NLRP3 inflammasome priming, licensing and assembly. In addition, we highlight the intricate and interconnected mechanisms involved in the activation of the NLRP3 inflammasome and the associated regulatory pathways. Furthermore, we discuss recent advances in the development of therapeutic strategies targeting the NLRP3 inflammasome to identify potential therapeutics for NLRP3-associated inflammatory diseases. As research continues to uncover the intricacies of the molecular networks governing NLRP3 activation, novel approaches for therapeutic interventions against NLRP3-related pathologies are emerging.
Collapse
Affiliation(s)
- Seungwha Paik
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- System Network Inflammation Control Research Center, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Biomedical Research Institute, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - Jin Kyung Kim
- Department of Microbiology, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Hyo Jung Shin
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Department of Biochemistry and Cell Biology, Eulji University School of Medicine, Daejeon, Republic of Korea
- Brain Research Institute, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Eun-Jin Park
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - In Soo Kim
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea
- Biomedical Research Institute, Chungnam National University Hospital, Daejeon, Republic of Korea
- Department of Pharmacology, Chungnam National University College of Medicine, Daejeon, Republic of Korea
| | - Eun-Kyeong Jo
- Department of Microbiology, Chungnam National University College of Medicine, Daejeon, Republic of Korea.
- Department of Medical Science, Chungnam National University College of Medicine, Daejeon, Republic of Korea.
- Biomedical Research Institute, Chungnam National University Hospital, Daejeon, Republic of Korea.
| |
Collapse
|
18
|
Park SK, Son JY, Kim YM, Ju JS, Ahn DK. BTX-A inhibited trigeminal neuralgia by blocking the NLRP3 pathway in rats. Brain Res Bull 2025; 225:111344. [PMID: 40220966 DOI: 10.1016/j.brainresbull.2025.111344] [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: 02/05/2025] [Revised: 04/02/2025] [Accepted: 04/09/2025] [Indexed: 04/14/2025]
Abstract
Few studies have examined the mechanisms underlying the development of trigeminal neuralgia involving the nucleotide-binding oligomerization domain, leucine-rich repeat, and pyrin domain-containing protein 3 (NLRP3). The purpose of this experiment was to investigate the role of NLRP3 in the antinociceptive effects of botulinum toxin type A (BTX-A) in trigeminal neuralgia. We used a trigeminal neuralgia animal model induced by injecting 1-acyl-2-lyso-sn-glycero-3-phosphate (LPA) into the trigeminal nerve root of rats. Rats treated with LPA showed a significant increase in the expression of NLRP3 in the trigeminal ganglion 9 days after LPA injection. Furthermore, the levels of interleukin (IL)-1β, IL-18, and tumor necrosis factor (TNF)-α increased on postoperative day 9. Subcutaneous administration of BTX-A (3 U/kg) in the vibrissa pad resulted in a significant attenuation of mechanical allodynia, and the antiallodynic effects lasted for 7 days. The upregulated NLRP3 expression in the trigeminal ganglion was suppressed 2 days after the injection of BTX-A. Moreover, the BTX-A injection significantly reduced the concentrations of IL-1β, IL-18, and TNF-α in the trigeminal ganglion. Intraganglionic injection of an NLRP3 inhibitor blocked mechanical allodynia and attenuated the upregulated cytokine concentrations in the LPA-treated rats. These results indicate that BTX-A induces its antinociceptive effects in the LPA-induced trigeminal neuralgia animal model by attenuating the NLRP3-cytokine pathway in the trigeminal ganglion.
Collapse
Affiliation(s)
- Se-Kyung Park
- Department of Oral Physiology, School of Dentistry and Craniofacial Nerve-Bone Network Research Center, Kyungpook National University, Daegu, Republic of Korea
| | - Jo-Young Son
- Department of Oral Physiology, School of Dentistry and Craniofacial Nerve-Bone Network Research Center, Kyungpook National University, Daegu, Republic of Korea
| | - Yu-Mi Kim
- Department of Oral Physiology, School of Dentistry and Craniofacial Nerve-Bone Network Research Center, Kyungpook National University, Daegu, Republic of Korea
| | - Jin-Sook Ju
- Department of Oral Physiology, School of Dentistry and Craniofacial Nerve-Bone Network Research Center, Kyungpook National University, Daegu, Republic of Korea
| | - Dong-Kuk Ahn
- Department of Oral Physiology, School of Dentistry and Craniofacial Nerve-Bone Network Research Center, Kyungpook National University, Daegu, Republic of Korea.
| |
Collapse
|
19
|
Li S, Huang Y, Sun Q, Li Y, Xie H, Fu Q. Caspase-1 is critical for mice in the defense against Streptococcus equi subsp. zooepidemicus infection by promoting macrophage phagocytosis. Microb Pathog 2025; 203:107499. [PMID: 40122410 DOI: 10.1016/j.micpath.2025.107499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 03/11/2025] [Accepted: 03/20/2025] [Indexed: 03/25/2025]
Abstract
Streptococcus equi subsp. zooepidemicus (SEZ) is an important pathogen which is responsible for a wide range of diseases in various species. Macrophages are professional phagocytes that can engulf microorganisms and trigger responses leading to microbial death. Caspase-1 is considered as a proinflammatory factor that mediates antibacterial response to protect hosts from bacteria. Here, we revealed a novel role of Caspase-1 in mice against SEZ. Through both in vitro and in vivo infection assays, we demonstrated that the maturation and secretion of the cytokine IL-1β are critically dependent on Caspase-1 activation. The Caspase-1 deficient mice displayed attenuation of bactericidal activity against SEZ, mainly by decreasing the accumulation of macrophage. In addition to the recruitment of macrophages, deficiency of Caspase-1 also impaired the phagocytosis of SEZ by macrophages. Our study demonstrated that Caspase-1 is critical for mice to defense against SEZ depending on the recruitment and phagocytosis of macrophage.
Collapse
Affiliation(s)
- Shun Li
- School of Animal Science and Technology, Foshan University, Guangdong, 528225, China; Foshan University Veterinary Teaching Hospital, Foshan University, Guangdong, 528225, China
| | - Yunfei Huang
- School of Animal Science and Technology, Foshan University, Guangdong, 528225, China; Foshan University Veterinary Teaching Hospital, Foshan University, Guangdong, 528225, China
| | - Qinqin Sun
- School of Animal Science and Technology, Foshan University, Guangdong, 528225, China; Foshan University Veterinary Teaching Hospital, Foshan University, Guangdong, 528225, China
| | - Yajuan Li
- School of Animal Science and Technology, Foshan University, Guangdong, 528225, China; Foshan University Veterinary Teaching Hospital, Foshan University, Guangdong, 528225, China
| | - Honglin Xie
- School of Animal Science and Technology, Foshan University, Guangdong, 528225, China; Foshan University Veterinary Teaching Hospital, Foshan University, Guangdong, 528225, China.
| | - Qiang Fu
- School of Animal Science and Technology, Foshan University, Guangdong, 528225, China; Foshan University Veterinary Teaching Hospital, Foshan University, Guangdong, 528225, China.
| |
Collapse
|
20
|
Wu K, Wang Q, Zhang Z, Luo W, Peng J, Ma X, Wang L, Xie C, Guo W. Honokiol ameliorates pyroptosis in intestinal ischemia‑reperfusion injury by regulating the SIRT3‑mediated NLRP3 inflammasome. Int J Mol Med 2025; 55:96. [PMID: 40280115 PMCID: PMC12045469 DOI: 10.3892/ijmm.2025.5537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 03/11/2025] [Indexed: 04/29/2025] Open
Abstract
Intestinal ischemia‑reperfusion (IIR) injury is caused by the restoration of blood supply after a period of ischemia. It occurs in numerous clinical pathologies, such as intestinal obstruction, incarcerated hernia and septic shock, with mortality rates of 50‑80%. Honokiol (HKL), isolated from the herb Magnolia officinalis, is a biphenolic natural product with antioxidative, antibacterial, antitumor and anti‑inflammatory properties. Additionally, HKL has protective effects in ischemia‑reperfusion injuries, but its role and specific mechanisms in IIR injury are yet to be elucidated. In the present study, the superior mesenteric artery was ligated in rats to establish an IIR model. Hematoxylin and eosin staining and ELISA revealed that HKL administration ameliorated IIR‑induced injury in rats, which was demonstrated by a reduced destruction to the intestinal mucosa, as well as a reduced serum intestinal fatty acid‑binding protein concentration and Chiu's score in 10 mg/kg HKL treated IIR‑induced rats compared with those without HKL treatment. Additionally, immunohistochemical (IHC) staining and western blotting revealed that the occludin and tight junction protein 1 protein levels were increased in the 10 mg/kg HKL treated IIR‑induced rats compared with those without HKL treatment. Furthermore, an in vitro hypoxia/reoxygenation (H/R) cell model was established using IEC‑6 cells. Cell Counting Kit‑8 and lactate dehydrogenase (LDH) assays indicated that HKL mitigated the H/R‑inhibited cell viability and decreased the LDH levels in cell supernatants. Mechanistically, immunofluorescent (IF) staining and western blotting revealed that HKL inhibited H/R‑triggered pyroptosis. Furthermore, Mito‑Tracker, mitochondrial membrane potential and MitoSOX staining as well as western blotting revealed that reducing mitochondrial reactive oxygen species (ROS) inhibited the H/R‑induced pyroptosis by mitigating mitochondrial dysfunction. In the present H/R cell model, HKL improved the mitochondrial function by increasing the expression of sirtuin 3 (SIRT3), while IF staining and western blotting indicated that silencing SIRT3 notably reduced the beneficial effect of HKL on pyroptosis. In addition, IHC staining and western blotting revealed that HKL treatment mitigated the IIR‑induced pyroptosis in rats. Therefore, HKL treatment may mitigate IIR‑induced mitochondrial dysfunction and reduce mitochondrial ROS production by increasing the expression of SIRT3 protein, potentially resulting in an inhibition of pyroptosis during IIR.
Collapse
Affiliation(s)
- Ke Wu
- Department of General Surgery, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Qiuling Wang
- Department of General Surgery, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Zhengyu Zhang
- Department of General Surgery, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Wei Luo
- Department of General Surgery, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Jing Peng
- Department of General Surgery, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Xin Ma
- Department of General Surgery, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Li Wang
- Research Center of Integrative Medicine, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Chunguang Xie
- Traditional Chinese Medicine Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610075, P.R. China
| | - Wubin Guo
- Department of General Surgery, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
- The Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Digestive System Diseases of Luzhou City, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| |
Collapse
|
21
|
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
|
22
|
Chen Y, Chen S, Liu Z, Wang Y, An N, Chen Y, Peng Y, Liu Z, Liu Q, Hu X. Red blood cells undergo lytic programmed cell death involving NLRP3. Cell 2025; 188:3013-3029.e19. [PMID: 40252640 DOI: 10.1016/j.cell.2025.03.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 08/27/2024] [Accepted: 03/24/2025] [Indexed: 04/21/2025]
Abstract
The canonical complement-mediated lysis of mature red blood cells (RBCs) leads to severe pathogenesis. However, inhibition strategies targeting complement are not always as efficient as expected, indicating that unknown mechanisms are awaiting elucidation. In this study, we investigate the intracellular events in mature RBCs following complement activation. The collected evidence demonstrates that complement-induced hemolysis is a caspase-8-dependent programmed RBC death. Furthermore, short NLRP3 (miniNLRP3) fragments in RBCs are identified to engage in the assembly of NLRP3-apoptosis-associated speck-like protein containing a CARD (ASC)-caspase-8 complex. Activated caspase-8 directly induces the proteolysis of β-spectrin, thereby disrupting the skeletal network of the RBC membrane, a process we refer to as spectosis. Spectosis signaling is also activated in autoimmune hemolytic anemia or paroxysmal nocturnal hemoglobinuria, and the inhibition of spectosis significantly reduced complement-induced hemolysis. These findings reveal a programmed death cascade in mature RBCs, which may have important implications for the treatment of hemolytic disorders.
Collapse
Affiliation(s)
- Yaozhen Chen
- Department of Transfusion Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, Shaanxi, China
| | - Shouwen Chen
- State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, East China University of Science and Technology, Shanghai 200237, China.
| | - Zhixin Liu
- Department of Transfusion Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, Shaanxi, China
| | - Yafen Wang
- Department of Transfusion Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, Shaanxi, China
| | - Ning An
- Department of Transfusion Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, Shaanxi, China
| | - Yutong Chen
- Department of Transfusion Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, Shaanxi, China
| | - Yihao Peng
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, Chinese University of Hong Kong, Shenzhen 518115, Guangdong, China
| | - Zheng Liu
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, Chinese University of Hong Kong, Shenzhen 518115, Guangdong, China
| | - Qin Liu
- State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, East China University of Science and Technology, Shanghai 200237, China.
| | - Xingbin Hu
- Department of Transfusion Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, Shaanxi, China.
| |
Collapse
|
23
|
Zhang Y, Chen M, Niu R, Guo D, Sun Z. Mechanistic Insights into T-2 Toxin-Induced Thymic Epithelial Cell Injury and Immunotoxicity via the ROS-NF-κB-NLRP3 Signaling Axis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:12961-12977. [PMID: 40387057 DOI: 10.1021/acs.jafc.5c00355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2025]
Abstract
Thymic epithelial cells (TECs) are critical for thymic structure and function, yet the impact of T-2 toxin (T-2) on TECs and related molecular pathways remains unclear. This study sheds light on the mechanisms of T-2-induced TEC damage, focusing on the ROS-NF-κB-NLRP3 signaling axis. The in vivo and in vitro analyses suggest that T-2 induces TEC injury through ROS-driven NLRP3 inflammasome activation, NF-κB signaling, inflammation, and apoptosis. Molecular docking analysis verified the binding of T-2 to critical components involved in oxidative stress, inflammatory signaling pathways, and apoptosis. These findings were further supported by therapeutic interventions targeting ROS and NLRP3. N-acetylcysteine (NAC) effectively reduced ROS levels, suppressed NF-κB signaling, inhibited NLRP3 activation, and mitigated inflammation and apoptosis, effects mirrored by the NLRP3 inhibitor MCC950, emphasizing the critical role of ROS-mediated NLRP3 inflammasome activation through NF-κB signaling in T-2-induced TEC damage. Concurrently, inhibition of the NF-κB signaling further suppressed ROS levels, NLRP3 inflammasome activation, and apoptosis in MTEC1 cells, emphasizing the pivotal function of the ROS-NF-κB-NLRP3 axis in the pathogenesis of T-2-induced thymic injury. Our study offers an in-depth insight into the mechanisms driving T-2-induced immunotoxicity and identifies potential therapeutic strategies targeting these pathways to mitigate thymic injury and preserve immune function.
Collapse
Affiliation(s)
- Yanfang Zhang
- School of Biological Engineering, Xinxiang University, Xinxiang, Henan 453000, China
| | - Mingyan Chen
- School of Biological Engineering, Xinxiang University, Xinxiang, Henan 453000, China
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, Shanxi 030801, China
| | - Ruiyan Niu
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, Shanxi 030801, China
| | - Dongguang Guo
- School of Biological Engineering, Xinxiang University, Xinxiang, Henan 453000, China
| | - Zilong Sun
- College of Veterinary Medicine, Shanxi Agricultural University, Jinzhong, Shanxi 030801, China
| |
Collapse
|
24
|
Abdelaziz AM. Alpha-Synuclein drives NURR1 and NLRP3 Inflammasome dysregulation in Parkinson's disease: From pathogenesis to potential therapeutic strategies. Int Immunopharmacol 2025; 156:114692. [PMID: 40267723 DOI: 10.1016/j.intimp.2025.114692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/10/2025] [Accepted: 04/17/2025] [Indexed: 04/25/2025]
Abstract
Parkinson's disease (PD), a progressive neurodegenerative disorder, is characterized by the loss of dopaminergic neurons and pathological aggregation of α-synuclein (α-Syn). Emerging evidence highlights the interplay between genetic susceptibility, neuroinflammation, and transcriptional dysregulation in driving PD pathogenesis. This review brings together the latest information on three important players: α-Syn, the transcription factor Orphan nuclear receptor (NURR1), and the NOD-like receptor 3 (NLRP3) inflammasome. Pathogenic α-syn aggregates cause damage to neurons by disrupting mitochondria and lysosomes and spreading in a way similar to prion proteins. They also turn on the NLRP3 inflammasome, which is a key player in neuroinflammation. NLRP3-driven release of pro-inflammatory cytokines exacerbates neurodegeneration and creates a self-sustaining inflammatory milieu. Meanwhile, reduced NURR1 activity, a pivotal modulator of dopaminergic neuron survival and development, exposes neurons to oxidative stress, neuroinflammation, and α-Syn toxicity, hence exacerbating disease progression. So, targeting this trio exhibits transformative potential against PD pathogenesis.
Collapse
Affiliation(s)
- Ahmed M Abdelaziz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Sinai University- Arish Branch, Arish 45511, Egypt.
| |
Collapse
|
25
|
Zheng L, Tang R, Ahmad F, Shi L, Chen X, Li J. hsa_circ_0081343 interacts with Rbm8a to inhibit NLRP3-mediated pyroptosis via the PI3K/AKT/HIF-1α pathways. Placenta 2025; 165:136-147. [PMID: 40267529 DOI: 10.1016/j.placenta.2025.04.005] [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: 12/23/2024] [Revised: 04/01/2025] [Accepted: 04/06/2025] [Indexed: 04/25/2025]
Abstract
INTRODUCTION Pyroptosis at the maternal-fetal interface plays an important role in fetal growth restriction development. hsa_circ_0081343 can be an RNA-binding protein "sponge" regulating Rbm8a nuclear transportation through binding to Rbm8a. This study aimed to elucidate the regulatory mechanism underlying the interaction between hsa_circ_0081343 and Rbm8a in the FGR pyroptosis pathway. METHODS The expression levels of PI3K/AKT pathway-related components (PI3K, AKT, p-PI3K, and p-AKT), HIF-1α, NLRP3, and proinflammatory cytokines (IL-1β, IL-6, and TNF-α) were measured using RT-qPCR, Western blot, and ELISA. RNA-seq and ChIP-seq were used to identify the downstream signaling pathways of hsa_circ_0081343 and Rbm8a in HTR8-SVneo. RNA pull-down assays, Western blot, and RT-qPCR were performed to investigate the interactions between hsa_circ_0081343 and Rbm8a. RESULT The placenta of FGR exhibited considerable upregulation of NLRP3 compared to normal controls. Overexpression of hsa_circ_0081343 inhibited pyroptosis and subsequent inflammatory responses in HTR-8/SVneo cells, and these effects were reversed by Rbm8a knockdown. The integration of RNA-seq and ChIP-seq showed that the PI3K/AKT and HIF-1α pathways were the targets of hsa_circ_0081343 and Rbm8a. hsa_circ_0081343 upregulation and Rbm8a downregulation was accompanied by the inhibition of the PI3K/AKT/HIF-1α signaling pathway, whereas hsa_circ_0081343 knockdown of and Rbm8a overexpression led to the opposite effect. Moreover, Rbm8a binds to hsa_circ_0081343, flanking the intron sequence. Rbm8a overexpression significantly decreased hsa_circ_0081343 expression. CONCLUSION These results indicated that the interaction between hsa_circ_0081343 and Rbm8a regulates NLRP3-mediated pyroptosis through the PI3K/AKT/HIF-1α signaling pathway. Furthermore, Rbm8a binds to hsa_circ_0081343, flanking the intron sequence and modulating hsa_circ_0081343 formation. Our results provide a new direction for further exploration of the regulatory mechanisms of circRNA-RBPs in the pathogenesis of FGR.
Collapse
Affiliation(s)
- Linmei Zheng
- Department of Obstetrics, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, China; Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Rong Tang
- Department of General Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, China
| | - Fiaz Ahmad
- Key Laboratory for Space Biosciences and Biotechnology, School of Life Sciences, Northwestern Polytechnical University (NPU), Xi'an, 710072, Shaanxi, China
| | - Lei Shi
- Department of Obstetrics, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, China
| | - Xiaoju Chen
- Department of Obstetrics, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, 570311, China.
| | - Jing Li
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
26
|
Moosavi Zenooz A, Eterafi M, Azarmi Giglou S, Safarzadeh E. Embracing cancer immunotherapy with manganese particles. Cell Oncol (Dordr) 2025:10.1007/s13402-025-01070-9. [PMID: 40397376 DOI: 10.1007/s13402-025-01070-9] [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: 02/15/2025] [Accepted: 05/05/2025] [Indexed: 05/22/2025] Open
Abstract
A substance integral to the sustenance and functionality of virtually all forms of life is manganese (Mn), classified as an essential trace metal. Its significance lies in its pivotal role in facilitating metabolic processes crucial for survival. Additionally, Mn exerts influence over various biological functions including bone formation and maintenance, as well as regulation within systems governing immunity, nervous signaling, and digestion. Manganese nanoparticles (Mn-NP) stand out as a beacon of promise within the realm of immunotherapy, their focus honed on intricate mechanisms such as triggering immune pathways, igniting inflammasomes, inducing immunogenic cell death (ICD), and sculpting the nuances of the tumor microenvironment. These minuscule marvels have dazzled researchers with their potential in reshaping the landscape of cancer immunotherapy - serving as potent vaccine enhancers, efficient drug couriers, and formidable allies when paired with immune checkpoint inhibitors (ICIs) or cutting-edge photodynamic/photothermal therapies. Herein, we aim to provide a comprehensive review of recent advances in the application of Mn and Mn-NP in the immunotherapy of cancer. We hope that this review will display an insightful view of Mn-NPs and provide guidance for design and application of them in immune-based cancer therapies.
Collapse
Affiliation(s)
- Ali Moosavi Zenooz
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
- Students Research Committee, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Majid Eterafi
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Soheil Azarmi Giglou
- Students Research Committee, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Elham Safarzadeh
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
- Department of Microbiology, Parasitology and Immunology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, 5166614711, Iran.
| |
Collapse
|
27
|
Jee YM, Lee JY, Ryu T. Chronic Inflammation and Immune Dysregulation in Metabolic-Dysfunction-Associated Steatotic Liver Disease Progression: From Steatosis to Hepatocellular Carcinoma. Biomedicines 2025; 13:1260. [PMID: 40427086 PMCID: PMC12109540 DOI: 10.3390/biomedicines13051260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2025] [Revised: 05/16/2025] [Accepted: 05/19/2025] [Indexed: 05/29/2025] Open
Abstract
Background/Objectives: Metabolic-dysfunction-associated steatotic liver disease (MASLD) progresses from hepatic steatosis to hepatocellular carcinoma (HCC) as a result of systemic immunometabolic dysfunction. This review summarizes the key roles of the innate and adaptive immune mechanisms driving hepatic injury, fibrogenesis, and carcinogenesis in MASLD. Methods: A comprehensive literature review was performed using PubMed to identify relevant published studies. Eligible articles included original research and clinical studies addressing immunological and metabolic mechanisms in MASLD, as well as emerging therapeutic strategies. Results: We highlight the roles of cytokine networks, the gut-liver axis, and immune cell reprogramming. Emerging therapeutic strategies, including cytokine inhibitors, anti-fibrotic agents, metabolic modulators, and nutraceuticals, offer several indications for attenuating MASLD progression and reducing the prevalence of extrahepatic manifestations. Conclusions: Given the heterogeneity of MASLD, personalized combination-based approaches targeting both inflammation and metabolic stress are essential for effective disease management and the prevention of systemic complications.
Collapse
Affiliation(s)
- Young-Min Jee
- Department of Family Medicine, Soonchunhyang University Seoul Hospital, Seoul 04401, Republic of Korea;
- Department of Family Medicine, Graduate School of Medicine, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Jeong-Yoon Lee
- Department of Neurology, Soonchunhyang University Seoul Hospital, Seoul 04401, Republic of Korea;
- Department of Translational Medicine, Graduate School of Medicine, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Tom Ryu
- Department of Internal Medicine, Institute for Digestive Research, Digestive Disease Center, Soonchunhyang University College of Medicine, Seoul 04401, Republic of Korea
| |
Collapse
|
28
|
Wadowski PP, Hülsmann M, Lang IM, Schörgenhofer C, Pultar J, Weikert C, Gremmel T, Steiner S, Koppensteiner R, Kopp CW, Jilma B. Glycocalyx Disintegration Is Associated with Mortality in Chronic Heart Failure. J Clin Med 2025; 14:3571. [PMID: 40429565 PMCID: PMC12112248 DOI: 10.3390/jcm14103571] [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: 04/02/2025] [Revised: 05/04/2025] [Accepted: 05/18/2025] [Indexed: 05/29/2025] Open
Abstract
Background: Glycocalyx disintegration is associated with adverse outcomes in patients with trauma or sepsis. As microvascular dysfunction has an impact on disease progression in chronic heart failure (CHF) patients, we hypothesized that changes in microcirculation might be associated with mortality. Methods: Fifty patients with ischemic and non-ischemic cardiomyopathy and conservative treatment with baseline measurements of the sublingual microcirculation (via Sidestream Darkfield videomicroscopy) were followed up for two years. Glycocalyx thickness was assessed indirectly by calculation of the perfused boundary region (PBR). Results: Loss of glycocalyx was pronounced in non-survivors after one, n = 10, and two years, n = 16; PBR: 2.05 μm (1.88-2.15 μm) vs. 1.87 μm (1.66-2.03 μm) and 2.04 (1.93-2.11) vs. 1.84 (1.62-1.97); p = 0.042 and p = 0.003, respectively. Area under the ROC curve for the analysis of the predictive value of PBR on two-year mortality was 0.77 (p = 0.003; SE: 0.07, CI (95%): 0.63-0.91). ROC curve analysis determined a PBR of 1.9 μm as the best predictor for two-year mortality (sensitivity: 0.81; specificity: 0.59). Moreover, multivariate regression analysis revealed PBR and functional capillary density as significant predictors of two-year mortality, p = 0.036 and p = 0.048, respectively. Conclusions: Glycocalyx disintegration is related to poor overall survival in CHF patients.
Collapse
Affiliation(s)
- Patricia P. Wadowski
- Department of Internal Medicine II, Division of Angiology, Medical University of Vienna, 1090 Vienna, Austria; (J.P.); (C.W.); (T.G.); (S.S.); (R.K.); (C.W.K.)
| | - Martin Hülsmann
- Department of Medicine II, Division of Cardiology, Medical University of Vienna, 1090 Vienna, Austria; (M.H.); (I.M.L.)
| | - Irene M. Lang
- Department of Medicine II, Division of Cardiology, Medical University of Vienna, 1090 Vienna, Austria; (M.H.); (I.M.L.)
| | - Christian Schörgenhofer
- Department of Clinical Pharmacology, Medical University of Vienna, 1090 Vienna, Austria; (C.S.)
| | - Joseph Pultar
- Department of Internal Medicine II, Division of Angiology, Medical University of Vienna, 1090 Vienna, Austria; (J.P.); (C.W.); (T.G.); (S.S.); (R.K.); (C.W.K.)
- Department of Anesthesia and Intensive Care Medicine, Universitätsklinikum St. Pölten, 3100 St. Pölten, Austria
| | - Constantin Weikert
- Department of Internal Medicine II, Division of Angiology, Medical University of Vienna, 1090 Vienna, Austria; (J.P.); (C.W.); (T.G.); (S.S.); (R.K.); (C.W.K.)
| | - Thomas Gremmel
- Department of Internal Medicine II, Division of Angiology, Medical University of Vienna, 1090 Vienna, Austria; (J.P.); (C.W.); (T.G.); (S.S.); (R.K.); (C.W.K.)
- Department of Internal Medicine I, Cardiology and Intensive Care Medicine, Landesklinikum Mistelbach-Gänserndorf, 2130 Mistelbach, Austria
- Institute of Cardiovascular Pharmacotherapy and Interventional Cardiology (Karl Landsteiner Society), 3100 St. Pölten, Austria
- Karl Landsteiner University of Health Sciences, 3500 Krems an der Donau, Austria
| | - Sabine Steiner
- Department of Internal Medicine II, Division of Angiology, Medical University of Vienna, 1090 Vienna, Austria; (J.P.); (C.W.); (T.G.); (S.S.); (R.K.); (C.W.K.)
| | - Renate Koppensteiner
- Department of Internal Medicine II, Division of Angiology, Medical University of Vienna, 1090 Vienna, Austria; (J.P.); (C.W.); (T.G.); (S.S.); (R.K.); (C.W.K.)
| | - Christoph W. Kopp
- Department of Internal Medicine II, Division of Angiology, Medical University of Vienna, 1090 Vienna, Austria; (J.P.); (C.W.); (T.G.); (S.S.); (R.K.); (C.W.K.)
| | - Bernd Jilma
- Department of Clinical Pharmacology, Medical University of Vienna, 1090 Vienna, Austria; (C.S.)
| |
Collapse
|
29
|
Zeng X, Yuan Y, Li Y, Hu Z, Hu S. Deciphering the NLRP3 inflammasome in diabetic encephalopathy: Molecular insights and emerging therapeutic targets. Exp Neurol 2025; 391:115304. [PMID: 40383363 DOI: 10.1016/j.expneurol.2025.115304] [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: 12/17/2024] [Revised: 05/01/2025] [Accepted: 05/11/2025] [Indexed: 05/20/2025]
Abstract
Diabetic encephalopathy (DE) is a neurological complication characterized by neuroinflammation, cognitive impairment, and memory decline, with its pathogenesis closely linked to the activation of the NLRP3 inflammasome. As a central regulator of the innate immune system, the NLRP3 inflammasome plays a pivotal role in DE progression by mediating neuroinflammation, pyroptosis, mitochondrial dysfunction, oxidative stress, endoplasmic reticulum (ER) stress, and microglial polarization. This review systematically explores the molecular mechanisms by which the NLRP3 inflammasome contributes to DE, focusing on its role in neuroinflammatory cascades and neuronal damage, as well as the diabetes-associated physiological changes that exacerbate DE pathogenesis. Furthermore, we summarize emerging therapeutic strategies targeting the NLRP3 inflammasome, including small-molecule inhibitors and bioactive compounds derived from traditional herbal medicine, highlighting their potential for DE treatment. These findings not only advance our understanding of DE but also provide a foundation for developing NLRP3-targeted pharmacological interventions.
Collapse
Affiliation(s)
- Xinyi Zeng
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330031, China; The First Clinical Medical College of Nanchang University, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330031, China
| | - Yi Yuan
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330031, China; School of Huankui Academy, Nanchang University, Nanchang, Jiangxi 330031, China
| | - Yujia Li
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330031, China; The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330031, China
| | - Ziyan Hu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330031, China; The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330031, China
| | - Shan Hu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330031, China.
| |
Collapse
|
30
|
Liu Y, Wu Q, Shao J, Mei Y, Zhang J, Xu Q, Mao L. The NLRP3 inflammasome: a therapeutic target of phytochemicals in treating atherosclerosis (a systematic review). Front Immunol 2025; 16:1568722. [PMID: 40443656 PMCID: PMC12119316 DOI: 10.3389/fimmu.2025.1568722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 04/22/2025] [Indexed: 06/02/2025] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease characterized by the gradual accumulation of plaques in arterial walls, with its pathogenesis remaining incompletely understood. Recent studies have highlighted that development of AS is closely associated with the aberrant activation of the NLRP3 inflammasome in the arteries. Inhibition of the NLRP3 inflammasome by natural products and formulae derived from Chinese herbal medicines (CHMs) has been shown to alleviate AS-associated pathologies. However, therapies that effectively and safely target the NLRP3 inflammasome remain limited. This review aims to summarize the key discoveries from recent studies on the effects of these natural products and formulae on the NLRP3 inflammasome in the context of AS treatment. A comprehensive literature search was conducted on databases such as PubMed/MEDLINE up to January 2025, yielding 38 eligible studies. Our analysis indicates that certain therapies can effectively prevent arterial inflammation in animal models by targeting multiple pathways and mechanisms related to the NLRP3 inflammasome. This review summarizes the primary findings of these studies, focusing on the therapeutic effects and underlying mechanisms of action. Based on these insights, we propose future strategies to enhance the efficacy, specificity, and safety of existing natural products and formulae for AS treatment. Additionally, this study offers a perspective for future research that may enhance our understanding of the roles and the mechanisms of CHM-derived phytochemicals and formulae in regulating the NLRP3 inflammasome and treating AS.
Collapse
Affiliation(s)
- Yongchao Liu
- Department of Immunology, School of Medicine, Nantong University, Nantong, China
| | - Qianyi Wu
- Department of Immunology, School of Medicine, Nantong University, Nantong, China
| | - Jing Shao
- Department of Immunology, School of Medicine, Nantong University, Nantong, China
| | - Youmin Mei
- Department of Periodontology, Nantong Stomatological Hospital, Nantong, China
| | - Jie Zhang
- Department of Immunology, School of Medicine, Nantong University, Nantong, China
| | - Qiuyun Xu
- Department of Immunology, School of Medicine, Nantong University, Nantong, China
| | - Liming Mao
- Basic Medical Research Center, School of Medicine, Nantong University, Nantong, China
| |
Collapse
|
31
|
Xu W, Wang L, Chen R, Liu Y, Chen W. Pyroptosis and its role in intestinal ischemia-reperfusion injury: a potential therapeutic target. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04261-1. [PMID: 40372474 DOI: 10.1007/s00210-025-04261-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Accepted: 05/02/2025] [Indexed: 05/16/2025]
Abstract
Intestinal ischemia-reperfusion injury (II/RI) is a critical acute condition characterized by complex pathological mechanisms, including various modes of cell death. Among these, pyroptosis has garnered significant attention in recent years. This review explores the characteristics, molecular mechanisms, and implications of pyroptosis in II/RI, with a focus on therapeutic strategies targeting the pyroptosis pathway. Key processes such as NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome activation, caspase-1 activation, and gasdermin D (GSDMD)-mediated membrane pore formation are identified as central to pyroptosis. Compounds like MCC950, CY-09, metformin, and curcumin have shown promise in attenuating II/RI in preclinical studies by modulating these pathways. However, challenges remain in understanding non-canonical pyroptosis pathways, unraveling the exact mechanisms of GSDMD-induced pore formation, and translating these findings into clinical applications. Addressing these gaps will be crucial for developing innovative and effective treatments for II/RI.
Collapse
Affiliation(s)
- Wenping Xu
- Department of Anesthesiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, 650032, China
| | - Lang Wang
- Department of Anesthesiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, 650032, China
| | - Ruili Chen
- Department of Anesthesiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, 650032, China
| | - Yi Liu
- Department of Anesthesiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, 650032, China
| | - Wendong Chen
- Department of Anesthesiology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, 650032, China.
| |
Collapse
|
32
|
Jiang B, Li XX, Lei Y, Wang XM, Wang TQ, Guo Z. Morphine exacerbates myocardial ischemia/reperfusion injury by overactivation of NLRP3 inflammasome via suppression of p-TRPV1 in male rats. Eur J Pharmacol 2025; 1000:177736. [PMID: 40381676 DOI: 10.1016/j.ejphar.2025.177736] [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: 02/24/2025] [Revised: 05/11/2025] [Accepted: 05/15/2025] [Indexed: 05/20/2025]
Abstract
The pathology of early MIRI (myocardial ischemia/reperfusion injury) is characterized by sterile inflammation. TRPV1 (transient receptor potential vanilloid 1) and NLRP3 inflammasome sense harmful stimulation and modulate inflammation in cardiomyocytes. We recently demonstrated morphine downregulated p-TRPV1 and exacerbated MIRI. In this study, we investigate the potential crosstalk of TRPV1 and NLRP3 inflammasome activities and a potential modulatory effect of morphine on the interaction in MIRI. In vivo and in vitro experiments were conducted. Coding RNA and pharmacological modulations were used in this study. We found MI/R (myocardial ischemia and reperfusion) upregulated p-TRPV1 without change in expression of NLRP3 inflammasome. Giving morphine during myocardial ischemia increased ventricular arrythmia, reduced heart rate and +dp/dt Max in reperfusion, and increased serum cTnI (cardiac troponin I) and infarct size. Suppression of p-TRPV1 but enhancement of NLRP3 inflammasome activity at the end of MI/R were detected. The alterations were reversed by an opioid μ-receptor antagonist or a NLRP3 inhibitor. Giving TRPV1 antagonist or knockdown of TRPV1 in cultured primary cardiomyocytes inhibited p-TRPV1 but increased NLRP3 inflammasome and the downstream cytokines and LDH (lactate dehydrogenase) in the supernatants. Conversely, treatment with capsaicin (a TRPV1 agonist) or upregulation of TRPV1 via transfection of Ad-TRPV1 elevated p-TRPV1 and reduced NLRP3 and LDH. The results indicated morphine treatment during myocardial ischemia aggravates MIRI by increasing the activity of NLRP3 inflammasome, via suppressing the inhibitory effect of p-TRPV1 on NLRP3 inflammasome. Targeting the signal chain of opioid μ-receptor agonist/TRPV1/NLRP3 inflammasome may find a novel way to improve MIRI.
Collapse
Affiliation(s)
- Bo Jiang
- College of Anesthesia, Shanxi Medical University, 86 Xinjiannan Road, Taiyuan, 030001, Shanxi, China
| | - Xiao-Xi Li
- College of Anesthesia, Shanxi Medical University, 86 Xinjiannan Road, Taiyuan, 030001, Shanxi, China; Department of Anesthesia, Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, China
| | - Yi Lei
- College of Anesthesia, Shanxi Medical University, 86 Xinjiannan Road, Taiyuan, 030001, Shanxi, China
| | - Xin-Meng Wang
- College of Anesthesia, Shanxi Medical University, 86 Xinjiannan Road, Taiyuan, 030001, Shanxi, China
| | - Tian-Qi Wang
- College of Anesthesia, Shanxi Medical University, 86 Xinjiannan Road, Taiyuan, 030001, Shanxi, China
| | - Zheng Guo
- College of Anesthesia, Shanxi Medical University, 86 Xinjiannan Road, Taiyuan, 030001, Shanxi, China; Department of Anesthesia, Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, China; Key Laboratory of Cellular Physiology (Shanxi Medical University), National Education Commission, Shanxi Medical University, 86 Xinjiannan Road, Taiyuan, 030001, Shanxi, China.
| |
Collapse
|
33
|
Yin J, Wan L, Zhang K, Yang J, Liu M, Zhao M, Li J. Progress of melatonin in the treatment of intervertebral disc degeneration. Front Physiol 2025; 16:1529315. [PMID: 40438255 PMCID: PMC12116318 DOI: 10.3389/fphys.2025.1529315] [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: 02/08/2025] [Accepted: 04/23/2025] [Indexed: 06/01/2025] Open
Abstract
The most common degenerative condition affecting the musculoskeletal system, and the leading cause of persistent low back pain, is intervertebral disc degeneration (IDD). IDD is increasingly common with age and has a variety of etiologic factors including inflammation, oxidative stress, extracellular matrix (ECM) degradation, and apoptosis that interact with each other to cause IDD. Because it is difficult to determine the exact pathogenesis of IDD, there is a lack of effective therapeutic agents. Melatonin has been intensively studied for its strong anti-inflammatory, antioxidant, and anti-apoptotic properties. Melatonin is a pleiotropic indole-stimulating hormone produced by the pineal gland, which can be used to treat a wide range of degenerative diseases. Therefore, melatonin supplementation may be a viable treatment for IDD. This article reviews the current mechanisms of IDD and the multiple roles regarding melatonin's anti-inflammatory, antioxidant, anti-apoptotic, and mitigating ECM degradation in IDD, incorporating new current research perspectives, as well as recent studies on drug delivery systems.
Collapse
Affiliation(s)
- Jianlin Yin
- Henan University of Chinese Medicine, Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital), Zhengzhou, China
| | - Lei Wan
- Henan University of Chinese Medicine, Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital), Zhengzhou, China
- Department of Osteology, The Second Affiliated Hospital of Luohe Medical College, Luohe, China
| | - Kuaixiang Zhang
- Henan University of Chinese Medicine, Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital), Zhengzhou, China
| | - Jiangjia Yang
- Graduate School, Hunan University of Chinese Medicine, Changsha, China
| | - Man Liu
- Henan University of Chinese Medicine, Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital), Zhengzhou, China
| | - Mingyu Zhao
- Henan University of Chinese Medicine, Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital), Zhengzhou, China
| | - Jitian Li
- Henan University of Chinese Medicine, Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital), Zhengzhou, China
- Graduate School, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
34
|
Li Y, Gao W, Qiu Y, Pan J, Guo Q, Liu X, Geng L, Shen Y, Deng Y, Hu Z, Li S, Liu S, Idris A, Huang J, Yang H, Ge B, Fan X, Chen X, Li J. RING1 dictates GSDMD-mediated inflammatory response and host susceptibility to pathogen infection. Cell Death Differ 2025:10.1038/s41418-025-01527-2. [PMID: 40369166 DOI: 10.1038/s41418-025-01527-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 04/27/2025] [Accepted: 05/06/2025] [Indexed: 05/16/2025] Open
Abstract
RING1 is an E3 ligase component of the polycomb repressive complex 1 (PRC1) with known roles in chromatin regulation and cellular processes such as apoptosis and autophagy. However, its involvement in inflammation and pyroptosis remains elusive. Here, we demonstrate that human RING1, not RING2, promotes K48-linked ubiquitination of Gasdermin D (GSDMD) and acts as a negative regulator of pyroptosis and bacterial infection. Indeed, we showed that loss of Ring1 increased S. typhimurium infectious load and mortality in vivo. Though RING1 deletion initially reduced M. tuberculosis (Mtb) infectious load in vivo, increased lung inflammation and impaired immune defense responses were later observed. Moreover, Ring1 knockout exacerbated acute sepsis induced by lipopolysaccharide (LPS) in vivo. Mechanistically, RING1 directly interacts with GSDMD and ubiquitinates the K51 and K168 sites of GSDMD for K48-linked proteasomal degradation, thereby inhibiting pyroptosis. Inhibition of RING1 E3 ligase activity by direct mutation or with the use of small molecule inhibitors increased GSDMD level and cell death during pyroptosis. Our findings reveal that RING1 dictates GSDMD-mediated inflammatory response and host susceptibility to pathogen infection, highlighting RING1 as a potential therapeutic target for combating infectious diseases.
Collapse
Affiliation(s)
- Yuanyuan Li
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Department of Neurology, Huashan Hospital and School of Life Sciences, Fudan University, Shanghai, China
| | - Wenqing Gao
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Department of Neurology, Huashan Hospital and School of Life Sciences, Fudan University, Shanghai, China
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Yuxin Qiu
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Department of Neurology, Huashan Hospital and School of Life Sciences, Fudan University, Shanghai, China
| | - Jiasong Pan
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Department of Neurology, Huashan Hospital and School of Life Sciences, Fudan University, Shanghai, China
| | - Qingqing Guo
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Department of Neurology, Huashan Hospital and School of Life Sciences, Fudan University, Shanghai, China
| | - Xuehe Liu
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Department of Neurology, Huashan Hospital and School of Life Sciences, Fudan University, Shanghai, China
| | - Lu Geng
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Department of Neurology, Huashan Hospital and School of Life Sciences, Fudan University, Shanghai, China
| | - Yajie Shen
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Department of Neurology, Huashan Hospital and School of Life Sciences, Fudan University, Shanghai, China
| | - Yifan Deng
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Department of Neurology, Huashan Hospital and School of Life Sciences, Fudan University, Shanghai, China
| | - Zhidong Hu
- Shanghai Institute of Infectious Diseases and Biosecurity & Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Suhua Li
- Division of Natural Science, Duke Kunshan University, Suzhou, China
| | - Shanshan Liu
- Shanghai Key Laboratory of Tuberculosis, Department of Microbiology and Immunology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Adi Idris
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Jinqing Huang
- Department of Chemistry, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Hua Yang
- Shanghai Key Laboratory of Tuberculosis, Department of Microbiology and Immunology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Baoxue Ge
- Shanghai Key Laboratory of Tuberculosis, Department of Microbiology and Immunology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaoyong Fan
- Shanghai Institute of Infectious Diseases and Biosecurity & Shanghai Public Health Clinical Center, Fudan University, Shanghai, China
| | - Xiangjun Chen
- Department of Neurology, Huashan Hospital and Institute of Neurology, Fudan University, Shanghai, China
| | - Jixi Li
- State Key Laboratory of Genetics and Development of Complex Phenotypes, Department of Neurology, Huashan Hospital and School of Life Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
35
|
Singh DD. NLRP3 inflammasome: structure, mechanism, drug-induced organ toxicity, therapeutic strategies, and future perspectives. RSC Med Chem 2025:d5md00167f. [PMID: 40370650 PMCID: PMC12070810 DOI: 10.1039/d5md00167f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Accepted: 04/22/2025] [Indexed: 05/16/2025] Open
Abstract
Drug-induced toxicity is an important issue in clinical medicine, which typically results in organ dysfunction and adverse health consequences. The family of NOD-like receptors (NLRs) includes intracellular proteins involved in recognizing pathogens and triggering innate immune responses, including the activation of the NLRP3 inflammasome. The NLRP3 (nucleotide-binding oligomerization domain-like receptor family, pyrin domain-containing 3) inflammasome is a critical component for both innate and adaptive immune responses and has been implicated in various drug-induced toxicities, including hepatic, renal, and cardiovascular diseases. The unusual activation of the NLRP3 inflammasome causes the release of pro-inflammatory cytokines, such as IL-1β and IL-18, which can lead to more damage to tissues. Targeting NLRP3 inflammasome is a potential therapeutic endeavour for suppressing drug-induced toxicity. This review provides insights into the mechanism, drug-induced organ toxicity, therapeutic strategies, and prospective therapeutic approaches of the NLRP3 inflammasome and summarizes the developing therapies that target the inflammasome unit. This review has taken up one of the foremost endeavours in understanding and inhibiting the NLRP3 inflammasome as a means of generating safer pharmacological therapies.
Collapse
Affiliation(s)
- Desh Deepak Singh
- Amity Institute of Biotechnology, Amity University Rajasthan Jaipur 303002 India +91 9450078260
| |
Collapse
|
36
|
Mao W, Liu X, Chen C, Luo T, Yan Z, Wu L, An Z. Roles for Exosomes from Various Cellular Sources in Spinal Cord Injury. Mol Neurobiol 2025:10.1007/s12035-025-05040-y. [PMID: 40347375 DOI: 10.1007/s12035-025-05040-y] [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: 02/29/2024] [Accepted: 05/04/2025] [Indexed: 05/12/2025]
Abstract
Spinal cord injury (SCI) is a severe disorder characterized by regeneration challenges in the central nervous system (CNS), resulting in permanent paralysis, loss of sensation, and abnormal autonomic functions. The complex pathophysiology of SCI poses challenges to traditional treatments, highlighting the urgent need for novel treatment approaches. Exosomes have emerged as promising candidates for SCI therapy because of their ability to deliver a wide range of bioactive molecules, such as RNAs, proteins, and lipids, to target cells with minimal immunogenicity, which contribute to anti-inflammatory, anti-apoptotic, autophagic, angiogenic, neurogenic, and axon remodeling activities. In this study, we classified exosomes from different sources into four categories based on the characteristics of the donor cells (mesenchymal stem cells, neurogenic cells, immune cells, vascular-associated cells) and provided a detailed summary and discussion of the current research progress and future directions for each source. We also conducted an in-depth investigation into the applications of engineered exosomes in SCI therapy, focusing on their roles in drug delivery and combination with surface engineering technologies and tissue engineering strategies. Finally, the challenges and prospects of exosomal clinical applications in SCI repair are described.
Collapse
Grants
- (No.202301039) Key Science and Technology planning project of Tongxiang City, Zhejiang Province, China
- (No.202301039) Key Science and Technology planning project of Tongxiang City, Zhejiang Province, China
- (No.202301039) Key Science and Technology planning project of Tongxiang City, Zhejiang Province, China
- (No.202301039) Key Science and Technology planning project of Tongxiang City, Zhejiang Province, China
- (No.202301039) Key Science and Technology planning project of Tongxiang City, Zhejiang Province, China
- (No.202301039) Key Science and Technology planning project of Tongxiang City, Zhejiang Province, China
- (No.202301039) Key Science and Technology planning project of Tongxiang City, Zhejiang Province, China
- (Zhejiang Health Commission Traditional Chinese Medicine [2019] No.1)). "13th Five-Year Plan" Traditional Chinese Medicine Key Specialty Construction Project of Zhejiang Province, China
- (Zhejiang Health Commission Traditional Chinese Medicine [2019] No.1)). "13th Five-Year Plan" Traditional Chinese Medicine Key Specialty Construction Project of Zhejiang Province, China
- (Zhejiang Health Commission Traditional Chinese Medicine [2019] No.1)). "13th Five-Year Plan" Traditional Chinese Medicine Key Specialty Construction Project of Zhejiang Province, China
- (Zhejiang Health Commission Traditional Chinese Medicine [2019] No.1)). "13th Five-Year Plan" Traditional Chinese Medicine Key Specialty Construction Project of Zhejiang Province, China
- (Zhejiang Health Commission Traditional Chinese Medicine [2019] No.1)). "13th Five-Year Plan" Traditional Chinese Medicine Key Specialty Construction Project of Zhejiang Province, China
- (Zhejiang Health Commission Traditional Chinese Medicine [2019] No.1)). "13th Five-Year Plan" Traditional Chinese Medicine Key Specialty Construction Project of Zhejiang Province, China
Collapse
Affiliation(s)
- Wangnan Mao
- The Second Clinical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xinghao Liu
- The Second Clinical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chen Chen
- Orthopedic Traumatology II, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Tongfu Luo
- The Second People's Hospital of Tongxiang City, Jiaxing, China
| | - Zheng Yan
- The Second Clinical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lianguo Wu
- Orthopedic Traumatology II, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.
| | - Zhongcheng An
- Orthopedic Traumatology II, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
37
|
Li M, Yin ZJ, Li L, Quan YY, Wang T, Zhu X, Tan RR, Zeng J, Hua H, Wu QX, Zhao JN. Rutaecarpine Attenuates Monosodium Urate Crystal-Induced Gouty Inflammation via Inhibition of TNFR-MAPK/NF-κB and NLRP3 Inflammasome Signaling Pathways. Chin J Integr Med 2025:10.1007/s11655-025-4204-3. [PMID: 40338445 DOI: 10.1007/s11655-025-4204-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/26/2024] [Indexed: 05/09/2025]
Abstract
OBJECTIVE To investigate the anti-inflammatory effect of rutaecarpine (RUT) on monosodium urate crystal (MSU)-induced murine peritonitis in mice and further explored the underlying mechanism of RUT in lipopolysaccharide (LPS)/MSU-induced gout model in vitro. METHODS In MSU-induced mice, 36 male C57BL/6 mice were randomly divided into 6 groups of 8 mice each group, including the control group, model group, RUT low-, medium-, and high-doses groups, and prednisone acetate group. The mice in each group were orally administered the corresponding drugs or vehicle once a day for 7 consecutive days. The gout inflammation model was established by intraperitoneal injection of MSU to evaluate the anti-gout inflammatory effects of RUT. Then the proinflammatory cytokines were measured by enzyme-linked immunosorbent assay (ELISA) and the proportions of infiltrating neutrophils cytokines were detected by flow cytometry. In LPS/MSU-treated or untreated THP-1 macrophages, cell viability was observed by cell counting kit 8 and proinflammatory cytokines were measured by ELISA. The percentage of pyroptotic cells were detected by flow cytometry. Respectively, the mRNA and protein levels were measured by real-time quantitative polymerase chain reaction (qRT-PCR) and Western blot, the nuclear translocation of nuclear factor κB (NF-κB) p65 was observed by laser confocal imaging. Additionally, surface plasmon resonance (SPR) and molecular docking were applied to validate the binding ability of RUT components to tumor necrosis factor α (TNF-α) targets. RESULTS RUT reduced the levels of infiltrating neutrophils and monocytes and decreased the levels of the proinflammatory cytokines interleukin 1β (IL-1β) and interleukin 6 (IL-6, all P<0.01). In vitro, RUT reduced the production of IL-1β, IL-6 and TNF-α. In addition, RT-PCR revealed the inhibitory effects of RUT on the mRNA levels of IL-1β, IL-6, cyclooxygenase-2 and TNF-α (P<0.05 or P<0.01). Mechanistically, RUT markedly reduced protein expressions of tumor necrosis factor receptor (TNFR), phospho-mitogen-activated protein kinase (p-MAPK), phospho-extracellular signal-regulated kinase, phospho-c-Jun N-terminal kinase, phospho-NF-κB, phospho-kinase α/β, NOD-like receptor thermal protein domain associated protein 3 (NLRPS), cleaved-cysteinyl aspartate specific proteinase-1 and cleaved-gasdermin D in macrophages (P<0.05 or P<0.01). Molecularly, SPR revealed that RUT bound to TNF-α with a calculated equilibrium dissociation constant of 31.7 µmol/L. Molecular docking further confirmed that RUT could interact directly with the TNF-α protein via hydrogen bonding, van der Waals interactions, and carbon-hydrogen bonding. CONCLUSION RUT alleviated MSU-induced peritonitis and inhibited the TNFR1-MAPK/NF-κB and NLRP3 inflammasome signaling pathway to attenuate gouty inflammation induced by LPS/MSU in THP-1 macrophages, suggesting that RUT could be a potential therapeutic candidate for gout.
Collapse
Affiliation(s)
- Min Li
- Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Zhu-Jun Yin
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, 610000, China
- The "Double-First Class" Application Characteristic Discipline of Hunan Province (Pharmaceutical Science), Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, 410219, China
| | - Li Li
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, 610000, China
| | - Yun-Yun Quan
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, 610000, China
| | - Ting Wang
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, 610000, China
| | - Xin Zhu
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, 610000, China
| | - Rui-Rong Tan
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, 610000, China
| | - Jin Zeng
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, 610000, China
| | - Hua Hua
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, 610000, China
| | - Qin-Xuan Wu
- The "Double-First Class" Application Characteristic Discipline of Hunan Province (Pharmaceutical Science), Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, 410219, China
| | - Jun-Ning Zhao
- Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.
- Translational Chinese Medicine Key Laboratory of Sichuan Province, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, 610000, China.
| |
Collapse
|
38
|
Wu J, Tang J, Huang D, Wang Y, Zhou E, Ru Q, Xu G, Chen L, Wu Y. Study on the comorbid mechanisms of sarcopenia and late-life depression. Behav Brain Res 2025; 485:115538. [PMID: 40122287 DOI: 10.1016/j.bbr.2025.115538] [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: 10/15/2024] [Revised: 03/06/2025] [Accepted: 03/09/2025] [Indexed: 03/25/2025]
Abstract
The increasing global aging population has brought greater focus to age-related diseases, particularly muscle-brain comorbidities such as sarcopenia and late-life depression. Sarcopenia, defined by the gradual loss of muscle mass and function, is notably prevalent among older individuals, while late-life depression profoundly affects their mental health and overall well-being. Epidemiological evidence suggests a high co-occurrence of these two conditions, although the precise biological mechanisms linking them remain inadequately understood. This review synthesizes the existing body of literature on sarcopenia and late-life depression, examining their definitions, prevalence, clinical presentations, and available treatments. The goal is to clarify the potential connections between these comorbidities and offer a theoretical framework for the development of future preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Jiale Wu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Jun Tang
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Di Huang
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Yu Wang
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Enyuan Zhou
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Qin Ru
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Guodong Xu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Lin Chen
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China.
| | - Yuxiang Wu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan 430056, China.
| |
Collapse
|
39
|
Xu M, Xu B. Protein lipidation in the tumor microenvironment: enzymology, signaling pathways, and therapeutics. Mol Cancer 2025; 24:138. [PMID: 40335986 PMCID: PMC12057185 DOI: 10.1186/s12943-025-02309-7] [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: 11/27/2024] [Accepted: 03/18/2025] [Indexed: 05/09/2025] Open
Abstract
Protein lipidation is a pivotal post-translational modification that increases protein hydrophobicity and influences their function, localization, and interaction network. Emerging evidence has shown significant roles of lipidation in the tumor microenvironment (TME). However, a comprehensive review of this topic is lacking. In this review, we present an integrated and in-depth literature review of protein lipidation in the context of the TME. Specifically, we focus on three major lipidation modifications: S-prenylation, S-palmitoylation, and N-myristoylation. We emphasize how these modifications affect oncogenic signaling pathways and the complex interplay between tumor cells and the surrounding stromal and immune cells. Furthermore, we explore the therapeutic potential of targeting lipidation mechanisms in cancer treatment and discuss prospects for developing novel anticancer strategies that disrupt lipidation-dependent signaling pathways. By bridging protein lipidation with the dynamics of the TME, our review provides novel insights into the complex relationship between them that drives tumor initiation and progression.
Collapse
Affiliation(s)
- Mengke Xu
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Intelligent Oncology Innovation Center Designated by the Ministry of Education, Chongqing University Cancer Hospital and Chongqing University School of Medicine, Chongqing, 400030, China
| | - Bo Xu
- Chongqing Key Laboratory of Intelligent Oncology for Breast Cancer, Intelligent Oncology Innovation Center Designated by the Ministry of Education, Chongqing University Cancer Hospital and Chongqing University School of Medicine, Chongqing, 400030, China.
| |
Collapse
|
40
|
Li S, Gong WL, Liu L, Shao B, Jiang SL, Li H, Song Y, Han GZ, Zhang ZQ. SiO 2 particles induce pulmonary fibrosis by modulating NLRP3 through the ROS/Keap1/Nrf2 signaling pathway in rats. Food Chem Toxicol 2025; 202:115520. [PMID: 40334973 DOI: 10.1016/j.fct.2025.115520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 04/28/2025] [Accepted: 05/05/2025] [Indexed: 05/09/2025]
Abstract
Recent studies have shown that the activation of the ROS-dependent NLRP3 inflammasome plays a key role in the pathogenesis of silicosis; however, the mechanism by which SiO2-induced ROS activates NLRP3 remains unclear. In this study, rats were intratracheally instilled with a SiO2 suspension once and then received daily intravenous injections of NAC (at doses of 20, 40, and 80 mg/kg, respectively) to inhibit SiO2-induced ROS. Rats that were intratracheally instilled with a SiO2 suspension once served as silicosis models, while those that were intratracheally instilled with PBS once served as controls. After 40 days, lung samples were taken for pathological observation, and the BALF was collected to measure ROS levels. The mRNA and protein expression levels of Keap1/Nrf2 signaling indicators (Keap1, Nrf2) and NLRP3 inflammasome indicators (NLRP3, GSDMD) were detected. The results showed that the Keap1/Nrf2 signaling pathway and the NLRP3 were activated in the silicosis rat lungs, accompanied by an increase in ROS levels. When ROS was inhibited, the Keap1/Nrf2 signaling pathway, the NLRP3, and the degree of pulmonary fibrosis were all suppressed in a dose-dependent manner. Therefore, we conclude that SiO2 particles induce pulmonary fibrosis in rats by modulating the NLRP3 inflammasome via the ROS/Keap1/Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Shuang Li
- Department of Public Health, Jining Medical University, Jining, China; Shandong Weixuankang Technology Innovation Co. LTD, Tai An, China
| | - Wei-Lei Gong
- Department of Public Health, Jining Medical University, Jining, China
| | - Lin Liu
- Health Management Center of Affiliated Hospital of Jining Medical University, Jining, China
| | - Bo Shao
- Department of Public Health, Jining Medical University, Jining, China
| | - Shun-Li Jiang
- Department of Public Health, Jining Medical University, Jining, China
| | - Huan Li
- Department of Public Health, Jining Medical University, Jining, China
| | - Ye Song
- Department of Public Health, Jining Medical University, Jining, China
| | - Gui-Zhi Han
- Department of Public Health, Jining Medical University, Jining, China.
| | - Zhao-Qiang Zhang
- Department of Public Health, Jining Medical University, Jining, China.
| |
Collapse
|
41
|
Huang C, Li J, Wu R, Li Y, Zhang C. Targeting pyroptosis for cancer immunotherapy: mechanistic insights and clinical perspectives. Mol Cancer 2025; 24:131. [PMID: 40319304 PMCID: PMC12049004 DOI: 10.1186/s12943-025-02344-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Accepted: 04/26/2025] [Indexed: 05/07/2025] Open
Abstract
Pyroptosis is a distinct form of programmed cell death characterized by the rupture of the cell membrane and robust inflammatory responses. Increasing evidence suggests that pyroptosis significantly affects the tumor microenvironment and antitumor immunity by releasing damage-associated molecular patterns (DAMPs) and pro-inflammatory mediators, thereby establishing it as a pivotal target in cancer immunotherapy. This review thoroughly explores the molecular mechanisms underlying pyroptosis, with a particular focus on inflammasome activation and the gasdermin family of proteins (GSDMs). It examines the role of pyroptotic cell death in reshaping the tumor immune microenvironment (TIME) involving both tumor and immune cells, and discusses recent advancements in targeting pyroptotic pathways through therapeutic strategies such as small molecule modulators, engineered nanocarriers, and combinatory treatments with immune checkpoint inhibitors. We also review recent advances and future directions in targeting pyroptosis to enhance tumor immunotherapy with immune checkpoint inhibitors, adoptive cell therapy, and tumor vaccines. This study suggested that targeting pyroptosis offers a promising avenue to amplify antitumor immune responses and surmount resistance to existing immunotherapies, potentially leading to more efficacious cancer treatments.
Collapse
Affiliation(s)
- Chen Huang
- Department of Biotherapy, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Jiayi Li
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Ruiyan Wu
- West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yangqian Li
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Chenliang Zhang
- Division of Abdominal Tumor Multimodality Treatment, Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
42
|
Pan L, Xie L, Yang W, Feng S, Mao W, Ye L, Cheng H, Wu X, Mao X. The role of brain-liver-gut Axis in neurological disorders. BURNS & TRAUMA 2025; 13:tkaf011. [PMID: 40321299 PMCID: PMC12048006 DOI: 10.1093/burnst/tkaf011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 02/04/2025] [Accepted: 02/07/2025] [Indexed: 05/08/2025]
Abstract
In recent years, with the increasing volume of related research, it has become apparent that the liver and gut play important roles in the pathogenesis of neurological disorders. Considering the interactions among the brain, liver, and gut, the brain-liver-gut axis has been proposed and gradually recognized. In this article, we summarized the complex network of interactions within the brain-liver-gut axis, encompassing the vagus nerve, barrier permeability, immunity and inflammation, the blood-brain barrier, gut microbial metabolites, the gut barrier, neurotoxic metabolites, and beta-amyloid (Aβ) metabolism. We also elaborated on the impact of the brain-liver-gut axis on various neurological disorders. Furthermore, we outline several therapies aimed at modulating the brain-liver-gut axis, including antibiotics, probiotics and prebiotics, fecal microbiota transplantation (FMT), vagus nerve stimulation (VNS), and dietary interventions. The focus is on elucidating possible mechanisms underlying neurological disorders pathogenesis and identifying effective treatments that are based on our understanding of the brain-liver-gut axis.
Collapse
Affiliation(s)
- Li Pan
- Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui 230022, China
| | - Lizheng Xie
- Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui 230022, China
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230022, China
| | - Wenpei Yang
- Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui 230022, China
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230022, China
| | - Shi Feng
- Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui 230022, China
| | - Wenbao Mao
- Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui 230022, China
| | - Lei Ye
- Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui 230022, China
| | - Hongwei Cheng
- Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui 230022, China
| | - Xiao Wu
- Department of Emergency, the First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui 230022, China
| | - Xiang Mao
- Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui 230022, China
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, No. 81 Meishan Road, Hefei, Anhui 230022, China
| |
Collapse
|
43
|
Gao K, Yang X, Zhao W, Lin Y, Hu B, Wang D. NAT10 PROMOTES PYROPTOSIS AND PANCREATIC INJURY OF SEVERE ACUTE PANCREATITIS THROUGH AC4C MODIFICATION OF NLRP3. Shock 2025; 63:774-780. [PMID: 39836947 DOI: 10.1097/shk.0000000000002551] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2025]
Abstract
ABSTRACT Severe acute pancreatitis (SAP) is a highly morbid acute digestive disorder linked to pyroptosis. N-acetyltransferase 10 (NAT10) facilitates the production of N4-acetylcytidine (ac4C) modifications in mRNA, thereby contributing to the progression of various diseases. However, the specific role of NAT10 in SAP remains to be elucidated. This study aimed to elucidate the mechanism through which NAT10 mediates pyroptosis in SAP. Sprague-Dawley rats and AR42J rat pancreatic exocrine cells were used to establish in vivo and in vitro models of SAP. The levels of ac4C and NAT10 expression were quantified using dot blot analysis and quantitative real-time PCR. Assessment of cell viability, apoptosis, amylase content, and concentrations of lactate dehydrogenase, IL-1β, and IL-18 was conducted to evaluate the severity of SAP both in vivo and in vitro . Pyroptosis was assessed by measuring caspase-1 and gasdermin D (GSDMD)-N-terminal (GSDMD-N) expression. Further mechanistic insights were gained using methylated RNA immunoprecipitation, RNA immunoprecipitation, and dual-luciferase reporter assays. Our findings indicate that the levels of ac4C modification and NAT10 were elevated in both in vivo and in vitro SAP models. Knockdown of NAT10 inhibited cell death and reduced the levels of amylase, lactate dehydrogenase, IL-1β, and IL-18 as well as the protein expression of caspase-1 and GSDMD-N, suggesting that NAT10 knockdown suppresses pyroptosis in SAP cell models. Mechanistically, NAT10 knockdown decreased the expression and stability of NOD-like receptor thermal protein domain-associated protein 3 (NLRP3) mRNA by inhibiting ac4C modification of NLRP3. Moreover, NAT10 knockdown alleviated pancreatic tissue pathology, mitigated SAP severity, and suppressed pyroptosis in an SAP rat model. Collectively, these results demonstrate that NAT10 exacerbates pancreatic injury in SAP by promoting pyroptosis through ac4C modification of NLRP3, thereby enhancing its expression. These findings suggest a potential novel therapeutic target for SAP.
Collapse
Affiliation(s)
- Ke Gao
- Department of Medical Oncology, The Forth Medical Center of PLA General Hospital, Beijing, China
| | - Xuejiao Yang
- Department of Medical Oncology, The Forth Medical Center of PLA General Hospital, Beijing, China
| | - Wei Zhao
- Senior Department of Hepato-Pancreato-Biliary Surgery, The First Medical Center of PLA General Hospital, Beijing, China
| | - Yipeng Lin
- Department of Hepatobiliary Surgery, Hainan Hospital of PLA General Hospital, Sanya, China
| | - Bin Hu
- Senior Department of Hepato-Pancreato-Biliary Surgery, The First Medical Center of PLA General Hospital, Beijing, China
| | - Dadong Wang
- Senior Department of Hepato-Pancreato-Biliary Surgery, The First Medical Center of PLA General Hospital, Beijing, China
| |
Collapse
|
44
|
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
|
45
|
Bu W, Yu M, Ma X, Shen Z, Ruan J, Qu Y, Huang R, Xue P, Ma Y, Tang J, Zhao X. Gender-specific effects of prenatal polystyrene nanoparticle exposure on offspring lung development. Toxicol Lett 2025; 407:1-16. [PMID: 40088994 DOI: 10.1016/j.toxlet.2025.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 02/21/2025] [Accepted: 03/08/2025] [Indexed: 03/17/2025]
Abstract
Nanoplastics are widely present in the environment. Exposure to environmental pollutants during pregnancy can have adverse effects on fetal development and health. Establishing a link between nanoplastics and Bronchopulmonary Dysplasia (BPD) requires further investigation. In this study, we examined the impact of prenatal exposure to 80 nm polystyrene nanoparticles (PS-NPs) on offspring lung development, taking into account potential gender-specific effects. Pregnant female mice were exposed to PS-NPs through oropharyngeal aspiration, and critical data on lung development were collected at postnatal days 1, 7, and 21. We found that exposure to PS-NPs reduced birth weight in female offspring and significantly increased lung weight in both male and female offspring by PND 21. Maternal exposure led to a reduction in alveolar numbers across offspring, with distinct underlying mechanisms observed between sexes. In female offspring, the reduction in alveolar numbers was linked to disrupted surfactant protein expression, significant inflammation, and increased apoptosis and fibrosis. In male offspring, impaired angiogenesis was the primary factor contributing to the increased risk of BPD. The impact on alveolar development was substantial in both genders. This study underscores the gender-specific impacts of prenatal nanoplastic exposure on lung development and offers new evidence and direction for future research on the cross-generational respiratory toxicity of PS-NPs.
Collapse
Affiliation(s)
- Wenxia Bu
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Mengjiao Yu
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Xinyi Ma
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Zhaoping Shen
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Jialing Ruan
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Yi Qu
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Ruiyao Huang
- Department of Clinical Medicine, Nantong University Xinglin College, Nantong 226000, China
| | - Peng Xue
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China
| | - Yuanyuan Ma
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China.
| | - Juan Tang
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China.
| | - Xinyuan Zhao
- Department of Occupational Medicine and Environmental Toxicology, Nantong Key Laboratory of Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China.
| |
Collapse
|
46
|
Yin WF, Wang MN, Zhang F, Pan XG, Ding K, Zhu H, Wang DG, Li LZ, Xia CY, Zhang WK, He J, Xu JK. Analgesic C 20-diterpenoid alkaloids from the lateral roots of Aconitum carmichaelii Debeaux: Natural inhibitors of NLRP3 activation. Bioorg Chem 2025; 158:108321. [PMID: 40080974 DOI: 10.1016/j.bioorg.2025.108321] [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: 12/18/2024] [Revised: 02/15/2025] [Accepted: 02/24/2025] [Indexed: 03/15/2025]
Abstract
Five new C20-diterpenoid alkaloids carmaloidlines A-E (1-5) and a known C20-diterpenoid alkaloid (6) were isolated from the lateral roots of Aconitum carmichaelii Debeaux. Their structures were determined through spectrometric analysis and quantum chemical calculations. Meanwhile, the plausible biosynthetic pathways of 1-6 were also discussed. Western blot results indicated that 2, 4, and 6 significantly inhibited caspase-1 maturation and IL-1β production on the NLRP3 signaling pathway at a concentration of 10 μM, among which 2 exhibited the greatest impact on NLRP3 inflammasome activation in a dose-dependent manner (5, 10 and 20 μM). Notably, the hot plate test in mice demonstrated that the analgesic efficacy of 2 was equivalent to that of morphine at the same dose (0.3 mg/kg), while also providing a longer pain latency period compared to morphine. Additionally, compound 2 effectively mitigated both mechanical allodynia and thermal hyperalgesia induced by the NLRP3 agonist nigericin at a dose of 0.03 mg/kg, exhibiting effects comparable to those of the NLRP3 inhibitor MCC950 (10 mg/kg). Molecular docking revealed that 2 could bind to the active site of MCC950 in NLRP3 protein, and 2 had the lower interaction energy. Cellular thermal shift assay (CETSA) further validated the binding of 2 to NLRP3. This could provide a scientific basis for developing NLRP3 activation inhibitors as novel analgesics.
Collapse
Affiliation(s)
- Wei-Feng Yin
- School of Life Sciences & School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, People's Republic of China; Institute of Clinical Medical Sciences & Department of Pharmacy, China-Japan Friendship Hospital, Beijing 100029, People's Republic of China
| | - Man-Ni Wang
- School of Life Sciences & School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, People's Republic of China
| | - Fan Zhang
- School of Life Sciences & School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, People's Republic of China
| | - Xue-Ge Pan
- School of Life Sciences & School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, People's Republic of China
| | - Kang Ding
- Institute of Clinical Medical Sciences & Department of Pharmacy, China-Japan Friendship Hospital, Beijing 100029, People's Republic of China
| | - Hai Zhu
- School of Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Dong-Ge Wang
- School of Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Ling-Zhi Li
- School of Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Cong-Yuan Xia
- Institute of Clinical Medical Sciences & Department of Pharmacy, China-Japan Friendship Hospital, Beijing 100029, People's Republic of China
| | - Wei-Ku Zhang
- Institute of Clinical Medical Sciences & Department of Pharmacy, China-Japan Friendship Hospital, Beijing 100029, People's Republic of China.
| | - Jun He
- Institute of Clinical Medical Sciences & Department of Pharmacy, China-Japan Friendship Hospital, Beijing 100029, People's Republic of China.
| | - Jie-Kun Xu
- School of Life Sciences & School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, People's Republic of China.
| |
Collapse
|
47
|
Tian Y, Li L, Sun Z, Liu J, Qiu C, Zhou J, Sun X, Lei Y. Decoding ozone's impact on the cornea: disruption of barrier integrity and its molecular drivers. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 296:118213. [PMID: 40267881 DOI: 10.1016/j.ecoenv.2025.118213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 03/30/2025] [Accepted: 04/15/2025] [Indexed: 04/25/2025]
Abstract
This study aims to investigate the influence of ozone exposure on mouse corneas and human corneal epithelial cells (HCEC) to better understand its impact on corneal health and the underlying molecular mechanisms. Elevated cyclic ozone exposure was applied to both mouse corneas and HCECs to assess its effects on corneal structure and cellular response. Ozone exposure induced corneal stromal thinning (27.88 %), increased epithelial thickness (22.44 %), and disrupted epithelial barrier function. Inflammatory responses and nitrative stress, marked by inflammatory cell infiltration and heightened 3-nitrotyrosine levels, coupled with the upregulation of NLRP3, caspase-1 were observed in mice cornea. Additionally, ozone exposure induced diminished cell viability, nitrative stress, and activation of the NLRP3/caspase-1/GSDMD pathway in HCECs, which were mitigated by anti-nitration agent MnTMPyP treatment. In summary, the study elucidated the mechanisms underlying ozone-induced corneal toxicity, highlighting nitrative stress and NLRP3 inflammasome-mediated pyroptosis. These findings suggest the importance of minimizing ozone exposure and also provide potential therapeutic strategies targeting nitrative stress and inflammasome activation to prevent ozone-related tissue damage.
Collapse
Affiliation(s)
- Yi Tian
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
| | - Liping Li
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
| | - Zhongmou Sun
- Bronxcare Health Systems, (EH), Bronx, NY, United States
| | - Jiamin Liu
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
| | - Chen Qiu
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
| | - Ji Zhou
- Shanghai Typhoon Institute, CMA, Shanghai 200030, China; Department of Atmospheric and Oceanic Sciences & Institute of Atmospheric Sciences, Fudan University, Shanghai 200031, China; Shanghai Key Laboratory of Meteorology and Health, Shanghai Meteorological Bureau, Shanghai 200030, China.
| | - Xinghuai Sun
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China; NHC Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, and Shanghai Key Laboratory of Visual Impairment and Restoration (Fudan University), Shanghai 200031, China; State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, Shanghai 200032, China.
| | - Yuan Lei
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, Shanghai 200031, China; NHC Key Laboratory of Myopia and Related Eye Diseases, Chinese Academy of Medical Sciences, and Shanghai Key Laboratory of Visual Impairment and Restoration (Fudan University), Shanghai 200031, China.
| |
Collapse
|
48
|
Gupta J, Mohammed MH, Alghazali T, Uthirapathy S, R R, Thakur V, Kaur M, Naidu KS, Kubaev A, Al-Mukhtar MM. Inflammasomes and autophagy in cancer: unlocking targeted therapies. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04184-x. [PMID: 40310530 DOI: 10.1007/s00210-025-04184-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Accepted: 04/13/2025] [Indexed: 05/02/2025]
Abstract
This study clarifies the interaction between autophagy and inflammasome within the cancer framework. The inflammasome generates pro-inflammatory cytokines to direct the immune response to pathogens and cellular stressors. Autophagy maintains cellular homeostasis and can either promote or inhibit cancer. These pathways interact to affect tumorigenesis, immune responses, and therapy. Autophagy controls inflammasome activity by affecting cancer pathogenesis and tumor microenvironment inflammation, highlighting novel cancer therapeutic approaches. Recent studies indicate that modulating autophagy and inflammasome pathways can boost anti-cancer immunity, reduce drug-resistance, and improve therapeutic efficacy. Recent studies indicate modulating inflammasome and autophagy pathways can augment anti-cancer immunity, mitigate therapy resistance, and improve treatment efficacy. Cancer research relies on understanding the inflammasome-autophagy relationship to develop targeted therapies that enhance anti-tumor efficacy and reduce inflammatory symptoms. Customized therapies may improve outcomes based on autophagy gene variations and inflammasome polymorphisms. This study investigates autophagy pathways and the inflammasome in tumor immunopathogenesis, cytokine function, and cancer therapeutic strategies, highlighting their significance in cancer biology and treatment.
Collapse
Affiliation(s)
- Jitendra Gupta
- Institute of Pharmaceutical Research, GLA University, Mathura, Pin Code 281406, U.P., India.
| | - Mohammed Hashim Mohammed
- Medical Laboratory Techniques Department, College of Health and Medical Technology, Al-Maarif University, Anbar, Iraq.
| | | | - Subasini Uthirapathy
- Pharmacy Department, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Roopashree R
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Vishal Thakur
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| | - Manpreet Kaur
- Department of Pharmacy, Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, 140307, Punjab, India
| | - K Satyam Naidu
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra, Pradesh- 531162, India
| | - Aziz Kubaev
- Department of Maxillofacial Surgery, Samarkand State Medical University, 18 Amir Temur Street, 140100, Samarkand, Uzbekistan
| | - Mahmoud Mussleh Al-Mukhtar
- Anesthesia Techniques Department, College of Health and Medical Techniques, Al-Mustaqbal University, 51001, Babylon, Iraq
| |
Collapse
|
49
|
Xiong L, Xiong Z, Hua J, Chen Q, Wang D. Mechanism of Nano-Microplastics Exposure-Induced Myocardial Fibrosis: DKK3-Mediated Mitophagy Dysfunction and Pyroptosis. J Biochem Mol Toxicol 2025; 39:e70245. [PMID: 40262053 DOI: 10.1002/jbt.70245] [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: 10/18/2024] [Revised: 02/25/2025] [Accepted: 03/18/2025] [Indexed: 04/24/2025]
Abstract
Nano-microplastics (NMPs), as environmental pollutants, are widely present in nature and pose potential threats to biological health. This study aims to investigate the mechanisms by which NMPs inhibit mitophagy through the suppression of dickkopf-related protein 3 (DKK3) expression, leading to NOD-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome-mediated cardiomyocyte pyroptosis and promoting myocardial fibrosis. Healthy adult male C57BL/6 mice were administered NMP solution via gavage, and their cardiac function was monitored. The results showed that NMP exposure significantly reduced left ventricular ejection fraction (LVEF) and left ventricular fractional shortening (LVFS) and increased the extent of myocardial fibrosis. Transcriptome sequencing identified 14 differentially expressed genes (DEGs), including MYL7. Using the random forest algorithm and functional enrichment analysis, DKK3 was identified as a key gene. In Vitro experiments further confirmed that NMPs downregulate DKK3 expression, thereby inhibiting mitophagy and promoting cardiomyocyte pyroptosis. This study elucidates the molecular mechanisms by which NMPs induce myocardial fibrosis and provides new theoretical bases and molecular targets for the diagnosis and treatment of heart diseases.
Collapse
Affiliation(s)
- Liang Xiong
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ziyi Xiong
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Juan Hua
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Qi Chen
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Dandan Wang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
50
|
El Hoss S, Shangaris P, Brewin J, Psychogyiou ME, Ng C, Pedler L, Rooks H, Gotardo ÉMF, Gushiken LFS, Brito PL, Nicolaides KH, Conran N, Rees DC, Strouboulis J. Reduced GATA1 levels are associated with ineffective erythropoiesis in sickle cell anemia. Haematologica 2025; 110:1150-1163. [PMID: 39633531 PMCID: PMC12050926 DOI: 10.3324/haematol.2024.286010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024] Open
Abstract
Ineffective erythropoiesis (IE) is defined as the abnormal differentiation and excessive destruction of erythroblasts i n the bone marrow, accompanied by an expanded progenitor compartment and relative reduction in the production of reticulocytes. It is a defining feature of many types of anemia, including β-thalassemia. GATA1 is an essential transcription factor for erythroid differentiation, known to be implicated in hematological conditions presenting with IE, including β-thalassemia and congenital dyserythropoietic anemia. However, little is known about the role of GATA1 in the erythropoietic defects recently described in sickle cell anemia (SCA). In the present study, we performed a detailed characterization of the role of GATA1 and ineffective erythropoiesis in SCA using both in vitro and in vivo assay systems. We demonstrate a significant decrease in GATA1 protein levels during SCA erythropoiesis and a concomitant increase in oxidative stress. Furthermore, we found that an increase in the activity of the inflammatory caspase, caspase 1, was driving the decrease in GATA1 levels during SCA erythropoiesis and that, upon inhibition of caspase 1 activity, SCA erythropoiesis was rescued and GATA1 levels partially restored. Our study further elucidates the defect in erythropoiesis in SCA, and may therefore help in the development of novel approaches to normalize the bone marrow niche prior to stem cell transplantation, or facilitate the production of healthy stem cells for gene therapy.
Collapse
Affiliation(s)
- Sara El Hoss
- Red Cell Haematology Lab, Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King's College London.
| | - Panicos Shangaris
- Women and Children's Health, School of Life Course and Population Sciences, King's College London, London, United Kingdom; Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom; Harris Birthright Research Centre for Fetal Medicine, King's College Hospital, London
| | - John Brewin
- Red Cell Haematology Lab, Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King's College London, United Kingdom; Department of Haematological Medicine, King's College Hospital, London
| | - Maria Eleni Psychogyiou
- Red Cell Haematology Lab, Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King's College London
| | - Cecilia Ng
- Red Cell Haematology Lab, Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King's College London
| | - Lauren Pedler
- Red Cell Haematology Lab, Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King's College London
| | - Helen Rooks
- Red Cell Haematology Lab, Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King's College London
| | - Érica M F Gotardo
- Hematology and Transfusion Center, Universidade Estadual de Campinas (UNICAMP), Campinas - São Paulo
| | - Lucas F S Gushiken
- Hematology and Transfusion Center, Universidade Estadual de Campinas (UNICAMP), Campinas - São Paulo
| | - Pâmela L Brito
- Hematology and Transfusion Center, Universidade Estadual de Campinas (UNICAMP), Campinas - São Paulo
| | - Kypros H Nicolaides
- Women and Children's Health, School of Life Course and Population Sciences, King's College London, London, United Kingdom; Harris Birthright Research Centre for Fetal Medicine, King's College Hospital, London
| | - Nicola Conran
- Hematology and Transfusion Center, Universidade Estadual de Campinas (UNICAMP), Campinas - São Paulo
| | - David C Rees
- Red Cell Haematology Lab, Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King's College London, United Kingdom; Department of Haematological Medicine, King's College Hospital, London
| | - John Strouboulis
- Red Cell Haematology Lab, Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King's College London.
| |
Collapse
|