1
|
Ahmed EI, Moneam Shamardl HA, Elsayed AM, Sadik SA. Evolocumab ameliorates myocardial fibrosis and improves metabolic syndrome-induced cardiac dysfunction in rats via inhibiting PCSK9/NLRP3 inflammasome and Caspase-1 / IL-1β pathways. Eur J Pharmacol 2025; 998:177499. [PMID: 40064223 DOI: 10.1016/j.ejphar.2025.177499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 02/19/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Affiliation(s)
- Eman Ibrahim Ahmed
- Pharmacology and Therapeutics Department, College of Medicine, Jouf University, Sakaka, Saudi Arabia; Medical Pharmacology Department, Faculty of Medicine, Fayoum University, Fayoum, Egypt.
| | | | - Asmaa Mohammed Elsayed
- Histology and Cell Biology Department, Faculty of Medicine, Fayoum University, Fayoum, Egypt
| | - Sawsan A Sadik
- Medical Pharmacology Department, Faculty of Medicine, Fayoum University, Fayoum, Egypt
| |
Collapse
|
2
|
Mo Y, Nie J, Zhang Y, Zhang Y, Yuan J, Zhang Q. HDAC6-Mediated NLRP3 Inflammasome Activation Is Involved in Nickel Nanoparticle-Induced Pulmonary Inflammation and Fibrosis. Chem Res Toxicol 2025; 38:877-891. [PMID: 40298147 DOI: 10.1021/acs.chemrestox.4c00551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Nickel nanoparticles (Nano-Ni) are increasingly utilized in industrial and biomedical applications, drawing growing attention to their potential adverse health effects. Our previous studies have demonstrated that Nano-Ni exposure induces severe, widespread, and persistent pulmonary inflammation and fibrosis. However, the underlying mechanisms are still unclear. The NLRP3 inflammasome is a vital component of the innate immune system and inflammatory signaling. In this study, we investigated whether Nano-Ni exposure activated the NLRP3 inflammasome and also examined its role in Nano-Ni-induced pulmonary inflammation and fibrosis. Our findings demonstrated that intratracheal instillation of wild-type mice (C57BL/6J) with 50 μg Nano-Ni per mouse resulted in NLRP3 inflammasome activation, IL-1β production, and extensive pulmonary inflammation and fibrosis. In contrast, Nano-Ni exposure induced only mild pulmonary inflammation and fibrosis in Nlrp3-/- mice (lacking functional NLRP3 inflammasome) or Il-1r1-/- mice (unresponsive to IL-1), highlighting the critical role of NLRP3 inflammasome activation in Nano-Ni-induced pulmonary damage. Further investigations using mouse alveolar macrophages (MH-S) revealed that Nano-Ni acts as a secondary activation signal for the NLRP3 inflammasome, triggering its activation in LPS-primed but not unprimed cells. Moreover, siRNA-mediated knockdown experiments demonstrated that this activation depended on Nano-Ni-induced upregulation of HDAC6. These findings suggest that Nano-Ni activates the NLRP3 inflammasome via HDAC6 as a second activation signal, leading to IL-1β production and subsequent pulmonary inflammation and fibrosis.
Collapse
Affiliation(s)
- Yiqun Mo
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, Louisville, Kentucky 40292, United States
| | - Jisheng Nie
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi 030001, China
- Key Laboratory of Coal Environmental Pathogenicity and Prevention (Shanxi Medical University), Ministry of Education, Taiyuan 030001, China
| | - Yue Zhang
- Department of Internal Medicine and Department of Dermatology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Yuanbao Zhang
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, Louisville, Kentucky 40292, United States
| | - Jiali Yuan
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, Louisville, Kentucky 40292, United States
| | - Qunwei Zhang
- Department of Epidemiology and Population Health, School of Public Health and Information Sciences, University of Louisville, Louisville, Kentucky 40292, United States
| |
Collapse
|
3
|
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
|
4
|
Mohamed MM, Zaki HF, Kamel AS. Possible Interaction of Suramin with Thalamic P2X Receptors and NLRP3 Inflammasome Activation Alleviates Reserpine-Induced Fibromyalgia-Like Symptoms. J Neuroimmune Pharmacol 2025; 20:51. [PMID: 40329125 PMCID: PMC12055955 DOI: 10.1007/s11481-025-10207-4] [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: 08/12/2024] [Accepted: 04/16/2025] [Indexed: 05/08/2025]
Abstract
The high pain sensitivity in fibromyalgia (FM) is processed by the thalamus that presents as a key component in the pain pathway in FM patients. Noteworthy, Purinergic receptors, specifically P2X, are implicated in pain signaling and neuroinflammation via inflammasome signaling. However, there is no available data on the impact of pharmacological intervention on the P2X receptor in thalamic pain transmission in FM. To investigate this aspect, the clinically tested P2X inhibitor, Suramin (SURM), was utilized. FM was induced over three days using Reserpine (1 mg/kg/day, s.c.), followed by a single dose of SURM (100 mg/kg, i.p.). At the molecular level, SURM countered the overexpression of P2X7 and P2X4 receptors accompanied by reduced NLRP3 inflammasome complex and pyroptotic markers like gasdermin-D. This was associated with the suppression of the p38-MAPK and NF-κB pathways, along with a decrease in pro-inflammatory cytokines and tumor necrosis factor-α as observed by increased CD86 expression on M1 microglia phenotype, a neuroinflammatory marker. Concurrently, blocking the P2X receptor shifted microglia polarization towards the M2 phenotype, marked by elevated CD163 expression, as a neuroprotective mechanism. This was outlined by increased neurotrophic and anti-inflammatory IL-10 with normalization of disturbed neurotransmitters. Behaviorally, SURM ameliorated the heightened pain processing, as observed in mechanical and thermal pain tests. Furthermore, it lowered Reserpine-induced motor impairment in the rotarod and open-field tests. This improvement in the somatosensory experience was reflected in alleviating depressive-like behavior in the forced swimming test. These findings highlight the therapeutic potential of blocking thalamic P2X receptors in alleviating fibromyalgia symptoms.
Collapse
Affiliation(s)
- Maram M Mohamed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Hala F Zaki
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Ahmed S Kamel
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| |
Collapse
|
5
|
Jung H, Kim B, Jang G, Kim H, Lee AR, Yoon SH, Lee KS, Hyun G, Kim Y, Ko J, Yu JW, Um JW. The NLRP3 inflammasome in microglia regulates repetitive behavior by modulating NMDA glutamate receptor functions. Cell Rep 2025; 44:115656. [PMID: 40333183 DOI: 10.1016/j.celrep.2025.115656] [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: 09/09/2024] [Revised: 02/21/2025] [Accepted: 04/15/2025] [Indexed: 05/09/2025] Open
Abstract
Neuroinflammation is a well-established risk factor for various neurological disorders and cognitive decline. However, the precise molecular mechanisms linking inflammation with neuropsychiatric symptoms remain unclear. Here, using NLRP3 (NOD-like receptor family, pyrin domain-containing protein 3) conditional knockin (cKI) mice harboring a D301N point mutation originating in patients with autoinflammatory diseases, we found that activation of the NLRP3 inflammasome by administration of lipopolysaccharide induced anxiety-like and repetitive behaviors frequently found in patients with neuropsychiatric disorders, as well as increasing NMDAR (N-methyl-D-aspartate receptor)-mediated excitatory synaptic functions in the medial prefrontal cortex of mice. In addition, interleukin 1β (IL-1β), a downstream cytokine of the NLRP3 inflammasome, enhanced NMDAR activation and increased surface levels of the selective NMDAR subunit GluN2A in cultured cortical neurons. Strikingly, treatment with an NMDAR antagonist or IL-1 receptor antagonist completely normalized the specific behavioral deficits in Nlrp3D301N-cKI mice. Collectively, our results demonstrate that NLRP3-mediated neuroinflammation elicits repetitive behavior through impaired NMDAR functions.
Collapse
Affiliation(s)
- Hyeji Jung
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu 42988, Korea; Center for Synapse Diversity and Specificity, DGIST, 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu 42988, Korea
| | - Byeongchan Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu 42988, Korea
| | - Gyubin Jang
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu 42988, Korea; Center for Synapse Diversity and Specificity, DGIST, 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu 42988, Korea
| | - Hyeonho Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu 42988, Korea; Center for Synapse Diversity and Specificity, DGIST, 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu 42988, Korea
| | - Ae-Ree Lee
- Core Protein Resources Center, DGIST, Daegu 42988, Korea; iProteinTherapeutics (iPT), Daegu 42988, Korea
| | - Sung-Hyun Yoon
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Kyung-Seo Lee
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Gaeun Hyun
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu 42988, Korea
| | - Younghye Kim
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu 42988, Korea; Center for Synapse Diversity and Specificity, DGIST, 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu 42988, Korea
| | - Jaewon Ko
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu 42988, Korea; Center for Synapse Diversity and Specificity, DGIST, 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu 42988, Korea
| | - Je-Wook Yu
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Ji Won Um
- Department of Brain Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu 42988, Korea; Center for Synapse Diversity and Specificity, DGIST, 333 Techno Jungangdae-Ro, Hyeonpoong-Eup, Dalseong-Gun, Daegu 42988, Korea.
| |
Collapse
|
6
|
Chen Q, Lai H, Chen Y, Peng Z, Wu S, Liu D. Characterization of circRNA expression profiles and functional roles in a mouse model of liver injury induced by OSA. Sci Rep 2025; 15:15615. [PMID: 40320447 PMCID: PMC12050294 DOI: 10.1038/s41598-025-99612-6] [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: 01/12/2025] [Accepted: 04/21/2025] [Indexed: 05/08/2025] Open
Abstract
Despite mounting evidence linking circular RNAs (circRNAs) to various diseases, their specific role in liver damage triggered by obstructive sleep apnea (OSA) remains ambiguous. This study investigates alterations in circRNA expression patterns in a mouse model subjected to chronic intermittent hypoxia (CIH), aiming to elucidate the pathways that lead to liver damage associated with OSA. We established the CIH model and conducted circRNA microarray analysis on liver samples from both CIH and control groups. The findings were substantiated via qRT-PCR. Furthermore, a comprehensive circRNA-miRNA-mRNA (ceRNA) network was developed, followed by the analysis of GO and KEGG pathways to further elucidate the underlying biological processes. We identified 259 differentially expressed circRNAs, comprising 86 that were upregulated and 173 that were downregulated in CIH mice. The ceRNA analysis suggested that these circRNAs may modulate gene expression by sequestering miRNAs. Our findings highlight potential therapeutic targets for liver pathologies associated with OSA.
Collapse
Affiliation(s)
- Qingshi Chen
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China.
| | - Huiting Lai
- Department of Interventional Therapy, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Yuwei Chen
- Department of Cardiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Zhuli Peng
- Department of Interventional Therapy, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Siying Wu
- Department of Interventional Therapy, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Dexin Liu
- Department of Interventional Therapy, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China.
| |
Collapse
|
7
|
Zhu Y, Zhao S, Tang Q, Xiong C, Chao Q, Huang L, Zhang J, Yao Y, Han G, Yang K. Screening and metabolic analysis of high-efficiency molluscicidal bacteria based on atmospheric and room temperature plasma mutagenesis. Acta Trop 2025; 265:107629. [PMID: 40280351 DOI: 10.1016/j.actatropica.2025.107629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 04/08/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
Oncomelania hupensis is the only intermediate host of Schistosoma japonicum, highlighting the need for developing low-toxicity, efficient, and economical molluscicides to control schistosomiasis transmission and prevalence. This study screened for effective molluscicides using O. hupensis in an immersion-based biological assay. Bsp dustable powder (DP) emerged as the most effective molluscicide among seven microbial pesticide samples tested. The dominant strain, designated 3-4, was isolated and identified as Bacillus subtilis using 16S rDNA gene sequencing. Following atmospheric and room temperature plasma (ARTP) mutagenesis, a mutant strain library containing 214 strains of bacteria was obtained. Most of the 139 mutant strains showed no significant difference compared with parental strain and 60 strains showed a decrease. Meanwhile, 15 mutant strains with higher molluscicidal effects were obtained. ARTP-129 and ARTP-154, exhibiting the highest positive mutation rates, demonstrated a 64.51 % increase in mortality compared to the parental strain 3-4. Metabolomic analysis revealed that the mutant strains may alter molluscicidal toxicity by regulating the synthesis pathways of metabolites such as l-pyroglutamic acid (PGA). These findings suggest the potential of ARTP mutagenesis for developing novel and effective molluscicides for schistosomiasis control.
Collapse
Affiliation(s)
- Yajing Zhu
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, PR China; Key Laboratory of National Health Commission on Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi, PR China
| | - Song Zhao
- Key Laboratory of National Health Commission on Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi, PR China; School of Biotechnology, Jiangnan University, Wuxi, Jiangsu, 214122, PR China
| | - Qianghui Tang
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, PR China; Key Laboratory of National Health Commission on Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi, PR China
| | - Chunrong Xiong
- Key Laboratory of National Health Commission on Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi, PR China
| | - Qiang Chao
- Key Laboratory of National Health Commission on Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi, PR China
| | - Lixin Huang
- National Agricultural Experimental Station for Agricultural Microbiology in Yangzhou, Lixiahe District Institute of Agricultural Sciences in Jiangsu, Yangzhou, PR China
| | - Jianfeng Zhang
- Key Laboratory of National Health Commission on Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi, PR China
| | - Yunyi Yao
- Key Laboratory of National Health Commission on Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi, PR China
| | - Guangjie Han
- National Agricultural Experimental Station for Agricultural Microbiology in Yangzhou, Lixiahe District Institute of Agricultural Sciences in Jiangsu, Yangzhou, PR China.
| | - Kun Yang
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, PR China; Key Laboratory of National Health Commission on Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi, PR China.
| |
Collapse
|
8
|
Li Z, Cheng W, Gao K, Liang S, Ke L, Wang M, Fan J, Li D, Zhang P, Xu Z, Li N. Pyroptosis: A spoiler of peaceful coexistence between cells in degenerative bone and joint diseases. J Adv Res 2025; 71:227-262. [PMID: 38876191 DOI: 10.1016/j.jare.2024.06.010] [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: 02/17/2024] [Revised: 05/23/2024] [Accepted: 06/07/2024] [Indexed: 06/16/2024] Open
Abstract
BACKGROUND As people age, degenerative bone and joint diseases (DBJDs) become more prevalent. When middle-aged and elderly people are diagnosed with one or more disorders such as osteoporosis (OP), osteoarthritis (OA), and intervertebral disc degeneration (IVDD), it often signals the onset of prolonged pain and reduced functionality. Chronic inflammation has been identified as the underlying cause of various degenerative diseases, including DBJDs. Recently, excessive activation of pyroptosis, a form of programed cell death (PCD) mediated by inflammasomes, has emerged as a primary driver of harmful chronic inflammation. Consequently, pyroptosis has become a potential target for preventing and treating DBJDs. AIM OF REVIEW This review explored the physiological and pathological roles of the pyroptosis pathway in bone and joint development and its relation to DBJDs. Meanwhile, it elaborated the molecular mechanisms of pyroptosis within individual cell types in the bone marrow and joints, as well as the interplay among different cell types in the context of DBJDs. Furthermore, this review presented the latest compelling evidence supporting the idea of regulating the pyroptosis pathway for DBJDs treatment, and discussed the potential, limitations, and challenges of various therapeutic strategies involving pyroptosis regulation. KEY SCIENTIFIC CONCEPTS OF REVIEW In summary, an interesting identity for the unregulated pyroptosis pathway in the context of DBJDs was proposed in this review, which was undertaken as a spoiler of peaceful coexistence between cells in a degenerative environment. Over the extended course of DBJDs, pyroptosis pathway perpetuated its activity through crosstalk among pyroptosis cascades in different cell types, thus exacerbating the inflammatory environment throughout the entire bone marrow and joint degeneration environment. Correspondingly, pyroptosis regulation therapy emerged as a promising option for clinical treatment of DBJDs.
Collapse
Affiliation(s)
- Zhichao Li
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Wenxiang Cheng
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Kuanhui Gao
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Songlin Liang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Liqing Ke
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Mengjie Wang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Jilin Fan
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Dandan Li
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang 050011, China
| | - Peng Zhang
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Faculty of Biomedical Engineering, Shenzhen University of Advanced Technology, Shenzhen 518000, China; Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, 518000 China; Shandong Zhongke Advanced Technology Co., Ltd., Jinan, 250300 China.
| | - Zhanwang Xu
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| | - Nianhu Li
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250014, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| |
Collapse
|
9
|
Tamer SA, Köse F, Yanar S, Budak Ö, Bağcı C. Anti-Inflammatory Effects of Spexin on Acetic Acid‑Induced Colitis in Rats via Modulating the NF-κB/NLRP3 Inflammasome Pathway. J Biochem Mol Toxicol 2025; 39:e70285. [PMID: 40320895 PMCID: PMC12050913 DOI: 10.1002/jbt.70285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2025] [Revised: 04/12/2025] [Accepted: 04/22/2025] [Indexed: 05/08/2025]
Abstract
Ulcerative colitis is a chronic inflammatory bowel disease characterized by inflammation and ulcers in the lining of the colon and rectum. Spexin is a novel peptide with antioxidant and anti-inflammatory properties. This study aims to elucidate the therapeutic effects and underlying mechanisms of spexin in mitigating acetic acid-induced colitis in rats. Male Sprague Dawley rats were assigned to control (n = 14) and colitis (n = 21) groups. Colitis was induced via 5% acetic acid (AA) administration (1 mL, intrarect). Post-induction, rats received subcutaneous saline (1 mL/kg), spexin (50 µg/kg/day), or oral sulfasalazine (500 mg/kg) for 5 days. Control groups received saline or spexin. After 24 h of the final treatment, colons were evaluated macroscopically, and levels of tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-18 were determined by ELISA, oxidative stress markers myeloperoxidase (MPO), malondialdehyde (MDA) and glutathione (GSH) levels were measured spectrophotometrically and NOD-like receptor pyrin domain-containing 3 (NLRP3), nuclear factor-κB (NF-κB), caspase-1 proteins were analyzed with Western Blot alongside histopathological assessments. Colitis induction significantly elevated macroscopic damage scores, stool consistency, inflammatory cytokines, MDA, MPO, and NLRP3, NF-κB, caspase-1, while reducing GSH levels (p < 0.001-0.01). Microscopic evaluations confirmed increased necrosis, submucosal edema, and inflammatory cell infiltration (p < 0.001). Spexin reversed these effects by enhancing GSH levels (p < 0.01), reducing macroscopic/microscopic scores, cytokines, MDA, and MPO levels (p < 0.05-0.001), and suppressing NLRP3, NF-κB, and caspase-1 activation (p < 0.01-0.001). For the first time that spexin ameluates acetic acid-induced colitis in rats by modulating the NF-κB/NLRP3 signaling pathway, reducing oxidative damage, enhancing antioxidant capacity, and suppressing inflammation.
Collapse
Affiliation(s)
- Sevil Arabacı Tamer
- Department of PhysiologySchool of Medicine, Sakarya UniversitySakaryaTürkiye
| | - Fadime Köse
- Department of PhysiologySchool of Medicine, Sakarya UniversitySakaryaTürkiye
| | - Sevinç Yanar
- Department of Histology and EmbryologySchool of Medicine, Sakarya UniversitySakaryaTürkiye
| | - Özcan Budak
- Department of Histology and EmbryologySchool of Medicine, Sakarya UniversitySakaryaTürkiye
| | - Cahit Bağcı
- Department of PhysiologySchool of Medicine, Sakarya UniversitySakaryaTürkiye
| |
Collapse
|
10
|
Ye L, Huang W, Li W, Yao Y, Peng Q, Fu Z, Xie S, He Q, Liu Y, Wan P, Sun B. Loteprednol etabonate alleviates NLRP3 inflammasome-associated inflammatory diseases in mice by suppressing the transcription of IL-1β. Int J Biol Macromol 2025; 306:141644. [PMID: 40032104 DOI: 10.1016/j.ijbiomac.2025.141644] [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: 01/02/2025] [Revised: 02/24/2025] [Accepted: 02/28/2025] [Indexed: 03/05/2025]
Abstract
Excessive activation of the NLRP3 inflammasome leads to cellular inflammation and tissue damage. Finding an inhibitor of its activation is urgent need for NLRP3 inflammasome-associated inflammatory diseases. In this study, we identified Loteprednol etabonate (LE), a well-known anti-inflammatory drug for ocular conditions, as a potent inhibitor of NLRP3 inflammasome activation through screening an FDA-approved drug library. In cellular models, LE significantly reduced IL-1β transcription, suppressed NLRP3 inflammasome activation, and finally inhibited the maturation and secretion of IL-1β and GSDMD-mediated pyroptosis. Mechanistic investigations showed that LE might inhibit IL-1β transcription by blocking both NF-κB and AP-1 signaling pathways. Furthermore, in mouse models of NLRP3 inflammasome-associated inflammatory diseases, including LPS-induced sepsis and DSS-induced colitis, intraperitoneal injection of LE significantly suppressed inflammatory response and improved mice survival rate. Collectively, these findings identify LE as a novel inhibitor of NLRP3 inflammasome activation, offering a promising therapeutic strategy for the treatment of NLRP3 inflammasome-associated inflammatory diseases.
Collapse
Affiliation(s)
- Lirui Ye
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China
| | - Weichen Huang
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China
| | - Weiling Li
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China
| | - Yulin Yao
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China
| | - Qian Peng
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China
| | - Zhengqi Fu
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China
| | - Shoufeng Xie
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China
| | - Qi He
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China
| | - Yuchen Liu
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China
| | - Pin Wan
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China; Department of Immunology, School of Medicine, Jianghan University, Wuhan 430056, China.
| | - Binlian Sun
- Hubei Key Laboratory of Cognitive and Affective Disorders, Institute of Biomedical Sciences, School of Medicine, Jianghan University, Wuhan 430056, China; Department of Immunology, School of Medicine, Jianghan University, Wuhan 430056, China.
| |
Collapse
|
11
|
Li T, Zhang Y, Li C, Song Y, Jiang T, Yin Y, Chang M, Song X, Zheng X, Zhang W, Yu Z, Feng W, Zhang Q, Ding L, Chen Y, Wang S. Microbial Photosynthetic Oxygenation and Radiotherapeutic Sensitization Enables Pyroptosis Induction for Combinatorial Cancer Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2503138. [PMID: 40285553 DOI: 10.1002/adma.202503138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 04/08/2025] [Indexed: 04/29/2025]
Abstract
Rectal cancer surgery is challenging due to the complex anatomy, making it difficult to achieve clear surgical margins. Radiotherapy (RT) plays a crucial role, especially in treating locally recurrent rectal cancer and preserving anal function. However, its effectiveness is often limited by tumor hypoxia, particularly prevalent in hypoxic regions near the bowel wall in colorectal cancer. Hypoxia contributes to both radiation resistance and apoptosis resistance, compromising RT outcomes. To overcome hypoxia-driven radiotherapy resistance, this work designs and engineers a radiotherapy-sensitizing bioplatform for efficient cancer RT. It combines lanthanum oxide nanoparticles (La2O3 NPs) with cyanobacteria, which produces oxygen through photosynthesis. This bioplatform uniquely reduces tumor hypoxia, enhances radiation deposition, and improves RT efficacy. La2O3 NPs further enhance reactive oxygen species (ROS) production induced by radiation, triggering pyroptosis via the ROS-NLRP3-GSDMD pathway, while RT amplifies pyroptosis through GSDME, circumventing tumor apoptosis resistance. The further integrated thermosensitive hydrogels ensure precise localization of the bioplatform, reducing systemic toxicity and improving therapeutic specificity. Compared to conventional therapies, this dual-action system addresses hypoxia, RT resistance, and apoptosis resistance more effectively. In vivo and in vitro hypoxia models validate its potent anti-tumor efficacy, offering valuable insights for refining clinical treatment paradigms.
Collapse
Affiliation(s)
- Tianyu Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, P. R. China
| | - Ya Zhang
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Cong Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, P. R. China
| | - Yanwei Song
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, P. R. China
| | - Tiaoyan Jiang
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Yipengchen Yin
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Meiqi Chang
- Laboratory Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, P. R. China
| | - Xinran Song
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Xiaojun Zheng
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Wenqing Zhang
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Zhongdan Yu
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Wei Feng
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Qin Zhang
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, P. R. China
| | - Li Ding
- Department of Orthopaedics, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Sheng Wang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, P. R. China
| |
Collapse
|
12
|
Li Z, Chen P, Qu A, Sun M, Xu L, Xu C, Hu S, Kuang H. Opportunities and Challenges for Nanomaterials as Vaccine Adjuvants. SMALL METHODS 2025:e2402059. [PMID: 40277301 DOI: 10.1002/smtd.202402059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 03/29/2025] [Indexed: 04/26/2025]
Abstract
Adjuvants, as a critical component of vaccines, are capable of eliciting more robust and sustained immune responses. Nanomaterials have shown unique advantages and broad application prospects in adjuvant development due to their high adjustability and distinctive physicochemical properties. This review focuses on nanoadjuvants and their immunological mechanisms. First, various types of adjuvants are introduced with an emphasis on metal and metal oxide nanoparticles, coordination polymers, liposomes, polymer nanoparticles, and other inorganic nanoparticles that can serve as vaccine adjuvants. Second, this review describes the current status of the clinical applications of nanoadjuvants. Next, the mechanisms of action for nanoadjuvants have been thoroughly elucidated, including the depot effect, NLRP3 inflammasome activation, targeting C-type lectin receptors, activation of toll-like receptors, and activation of the cGAS-STING signaling pathway. Finally, the challenges and opportunities associated with the development of nanoadjuvants have also been addressed.
Collapse
Affiliation(s)
- Zongda Li
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Panpan Chen
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Aihua Qu
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Maozhong Sun
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Liguang Xu
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Chuanlai Xu
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Shudong Hu
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| | - Hua Kuang
- International Joint Research Laboratory for Biointerface and Biodetection, State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, 214122, China
| |
Collapse
|
13
|
Xu H, Xu H, Li W, Liang Z, Luo W, Sheng S, Liang G, Zhang Z. Modulating the NLRP3 Inflammasome: Acitretin as a potential treatment for Sepsis-induced acute lung injury. Int Immunopharmacol 2025; 153:114504. [PMID: 40187888 DOI: 10.1016/j.intimp.2025.114504] [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: 01/27/2025] [Revised: 03/03/2025] [Accepted: 03/15/2025] [Indexed: 04/07/2025]
Abstract
BACKGROUND Acitretin, a well-established dermatological drug primarily used for psoriasis treatment, has been clinically used for several decades. However, its potential role in modulating inflammation in sepsis remains unexplored. OBJECTIVE This study seeks to explore the impact of acitretin on sepsis-induced acute lung injury (ALI) and to elucidate the underlying mechanisms involved. METHODS In a mouse model of sepsis induced by lipopolysaccharide (LPS), we assessed the effects of acitretin on ALI. Transcriptome sequencing of lung tissue was performed to identify relevant signaling pathways. In vitro, bone marrow-derived macrophages (BMDMs) were treated with acitretin (1 μM, 5 μM and 10 μM) to evaluate its impact on NOD-, LRR- and pyrin domain-containing protein 3(NLRP3) inflammasome activation and pyroptosis. In vivo, wild-type, Nlrp3 knockout, and Gsdmd knockout mice were used to confirm the role of the NLRP3 inflammasome in mediating acitretin's effects. RESULTS Acitretin significantly mitigated sepsis-induced ALI, reducing mortality in LPS-challenged mice. Transcriptome analysis revealed that acitretin suppressed the NLRP3 inflammasome pathway in lung tissue. In vitro, acitretin dose-dependently inhibited interleukin (IL)-1β release, caspase-1 p20 production, and GSDMD cleavage in BMDMs. Furthermore, acitretin inhibited inflammasome activation by preventing ASC oligomerization and its interaction with NLRP3. In vivo, acitretin reduced lung tissue inflammation, IL-1β levels in bronchoalveolar lavage fluid, and the ratio of wet to dry in wide-type mice, but these effects were abolished in Nlrp3 and Gsdmd knockout mice. CONCLUSION Acitretin demonstrated significant anti-inflammatory properties through the suppression of the NLRP3 inflammasome, suggesting its potential as a therapeutic strategy for sepsis and related complications.
Collapse
Affiliation(s)
- Huikang Xu
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Haowen Xu
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Weifeng Li
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou 311399, China
| | - Zhiyu Liang
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou 311399, China
| | - Weiwei Luo
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Shiying Sheng
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Guang Liang
- School of Pharmaceutical Sciences, Hangzhou Medical College, Hangzhou 311399, China.
| | - Zhaocai Zhang
- Department of Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Key Laboratory of the Diagnosis and Treatment for Severe Trauma and Burn of Zhejiang Province, Hangzhou, China; Zhejiang Province Clinical Research Center for Emergency and Critical Care Medicine, Hangzhou, China.
| |
Collapse
|
14
|
Yang C, Wang L, Liu Y, Zhang Y, Jin C, Cheng J, Shang L, Fang L, Wu S, Chen C, Wang J. Thermal Proteome Profiling Reveals Meltome Upon NLRP3 Inflammasome Activation. Mol Cell Proteomics 2025; 24:100972. [PMID: 40250624 DOI: 10.1016/j.mcpro.2025.100972] [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: 02/06/2025] [Revised: 03/31/2025] [Accepted: 04/14/2025] [Indexed: 04/20/2025] Open
Abstract
NOD-like receptor (NLR) family pyrin domain containing 3 (NLRP3) involves in inflammasome complex assembly and innate immunity. Activation of the NLRP3 inflammasome induces conformational alterations in protein complexes, influencing their interactions with other molecules, which in turn affects protein thermal stability. To investigate the proteome-wide thermal stability alterations induced by NLRP3 inflammasome activation, we conducted a comprehensive analysis of meltome dynamics using thermal proteome profiling. Our analysis identified 337 proteins exhibiting alterations in thermal stability upon NLRP3 inflammasome activation. Subsequently, we validated three proteins by the cellular thermal shift assay. Notably, our findings reveal that the majority of these proteins tend to cluster into distinct macromolecular complexes. Furthermore, we identified FAM120A as a novel NLRP3 binding partner, with its suppression enhancing caspase-1 activation and IL-1β release in response to NLRP3 agonist. Collectively, these data provide a comprehensive framework for understanding the mechanisms of NLRP3 inflammasome activation and underscore the utility of thermal proteome profiling in exploring proteome-wide thermal stability changes during signaling transduction.
Collapse
Affiliation(s)
- Chen Yang
- College of Life Sciences, Hebei University, Baoding, China; State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Ling Wang
- College of Life Sciences, Hebei University, Baoding, China; State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Yuchen Liu
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Yuehui Zhang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Chaozhi Jin
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Jiale Cheng
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China; School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Limin Shang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Longlong Fang
- College of Life Sciences, Hebei University, Baoding, China; State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Shanshan Wu
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Chuan Chen
- College of Life Sciences, Hebei University, Baoding, China
| | - Jian Wang
- College of Life Sciences, Hebei University, Baoding, China; State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China.
| |
Collapse
|
15
|
Cui X, Qiao R, Wang B, Hu Y, Sun G, Hu W, Luan Z, Ren H, Xu H, Guan Y, Zhang X. Uric acid reduces the expression of aquaporins in renal collecting ducts to increase urine output in hyperuricemia. Front Physiol 2025; 16:1504328. [PMID: 40271210 PMCID: PMC12014756 DOI: 10.3389/fphys.2025.1504328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 03/13/2025] [Indexed: 04/25/2025] Open
Abstract
Background Hyperuricemia (HUA) has attracted wide attention due to its close relationship with gout, hypertension, hypertriglyceridemia, obesity, atherosclerotic heart disease, type 2 diabetes and chronic kidney disease. Clinical observations suggest that people with high levels of serum uric acid (sUA) exhibits impaired urine concentration. We speculate that UA may regulate the expression of AQPs through inflammatory pathways, resulting in impaired renal urine concentration. Methods and results We revealed that patients and mice with HUA had a polyuria phenotype and found that the expression of aquaporin 2 (AQP2), AQP3 and AQP4 were significantly reduced in the kidneys of mice with HUA. Similarly, uric acid (UA) treatment markedly suppressed the expression of AQP2, AQP3 and AQP4 in cultured inner medullary collecting duct cells (IMCDs). We observed an increased expression of NF-κB in the kidneys of mice with HUA and in the IMCD cells treated with UA. Blockade of NF-κB by its inhibitor Bay 11-7082 dramatically attenuated UA-suppressed expression of AQP2, AQP3 and AQP4. Furthermore, the luciferase reporter, CHIP and EMSA assays showed that NF-κB can directly bind to the promoter regions of AQP2, AQP3 and AQP4 genes to suppress their transcription. Conclusion Our findings demonstrate that UA reduces the expression of AQP2, AQP3 and AQP4 in an NFκB-dependent manner, which contributes to the polyuria phenotype in the subjects with HUA.
Collapse
Affiliation(s)
- Xiaohui Cui
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Rongfang Qiao
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Bing Wang
- Department of Endocrinology and Metabolism, The Central hospital of Dalian University of Technology, Dalian, China
| | - Yitong Hu
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Guoying Sun
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Wenjuan Hu
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Zhilin Luan
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Huiwen Ren
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Hu Xu
- Kidney Health Institute, East China Normal University, Shanghai, China
| | - Youfei Guan
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Xiaoyan Zhang
- Kidney Health Institute, East China Normal University, Shanghai, China
| |
Collapse
|
16
|
Hsu CW, Okano T, Niinuma Y, Leewananthawet A, Iida T, Onsoi P, Boonyaleka K, Ashida H, Suzuki T. A complex of NLRP3 with caspase-4 is essential for inflammasome activation by Tannerella forsythia infection. Int Immunol 2025; 37:261-271. [PMID: 39673522 DOI: 10.1093/intimm/dxae071] [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: 09/24/2024] [Accepted: 12/02/2024] [Indexed: 12/16/2024] Open
Abstract
Periodontitis, a chronic inflammatory disease of periodontal tissue, is often associated with a group of pathogenic bacteria known as the "red complex", including Tannerella forsythia. Previous papers showed that T. forsythia induces many kinds of inflammatory cytokines including interleukin (IL)-1β regulated by inflammasome activation. However, the physiological function of periodontitis and the mechanism to induce inflammasome activation by T. forsythia infection are poorly understood. In this study, we demonstrate that the Nod-like receptor pyrin domain containing 3 (NLRP3) and caspase-4 are essential for inflammasome activation by T. forsythia infection, playing a crucial role in IL-1β maturation in THP-1 cells. We also showed that the knockout of ASC or Gasdermin D suppresses pyroptotic cell death. Moreover, co-immunoprecipitation assays confirmed the formation of a complex involving caspase-4, NLRP3, and ASC following T. forsythia infection. Additionally, reactive oxygen species production was identified as a key factor in caspase-4-mediated NLRP3 inflammasome activation by T. forsythia infection. These results enhance our understanding of inflammasome activation in response to T. forsythia infection and provide new insights into the pathogenic mechanisms of periodontitis.
Collapse
Affiliation(s)
- Chen-Wei Hsu
- Department of Bacterial Pathogenesis, Infection and Host Response, Graduate School of Medical and Dental Sciences, Institute of SCIENCE TOKYO, Tokyo, Japan
| | - Tokuju Okano
- Department of Bacterial Pathogenesis, Infection and Host Response, Graduate School of Medical and Dental Sciences, Institute of SCIENCE TOKYO, Tokyo, Japan
| | - Yuiko Niinuma
- Department of Bacterial Pathogenesis, Infection and Host Response, Graduate School of Medical and Dental Sciences, Institute of SCIENCE TOKYO, Tokyo, Japan
| | - Anongwee Leewananthawet
- Specialized Dental Center, Department of Restorative Dentistry and Periodontology, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| | - Tamako Iida
- Department of Bacterial Pathogenesis, Infection and Host Response, Graduate School of Medical and Dental Sciences, Institute of SCIENCE TOKYO, Tokyo, Japan
| | - Poramed Onsoi
- Department of Bacterial Pathogenesis, Infection and Host Response, Graduate School of Medical and Dental Sciences, Institute of SCIENCE TOKYO, Tokyo, Japan
| | - Kotchakorn Boonyaleka
- Division of Periodontology, Department of Restorative Dentistry and Periodontology, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| | - Hiroshi Ashida
- Department of Bacterial Pathogenesis, Infection and Host Response, Graduate School of Medical and Dental Sciences, Institute of SCIENCE TOKYO, Tokyo, Japan
| | - Toshihiko Suzuki
- Department of Bacterial Pathogenesis, Infection and Host Response, Graduate School of Medical and Dental Sciences, Institute of SCIENCE TOKYO, Tokyo, Japan
| |
Collapse
|
17
|
Huang Z, Chen G, Ren Z, Xiao L, Chen Z, Xie Y, Wang G, Zhou B. Urolithin A ameliorates schizophrenia-like behaviors and cognitive impairments in female rats by modulating NLRP3 signaling. Int Immunopharmacol 2025; 151:114336. [PMID: 39987632 DOI: 10.1016/j.intimp.2025.114336] [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: 09/19/2024] [Revised: 02/16/2025] [Accepted: 02/16/2025] [Indexed: 02/25/2025]
Abstract
The management of cognitive impairments in schizophrenia presents a considerable challenge, with a strong association between neuroinflammation and its progression. Urolithin A (UA) demonstrates important anti-inflammatory properties in multiple neurological disease models, contributing to the enhancement of cognitive deficits. However, it remains uncertain if UA can produce comparable neuroregulatory effects in female rat models of schizophrenia. Eight-week-old female Sprague Dawley rats received either 0.1 mg/kg of MK801 or volume-matched saline via intraperitoneal injection for 5 consecutive days. Furthermore, they were administered 150 mg/kg of UA through oral gavage for 4 weeks. Behavioral assessments were performed to evaluate cognitive function and behavior after UA treatment. Immunofluorescence staining was employed to assess microglial activity in the hippocampus, while Western blot analysis was conducted to investigate the expression of neuroinflammation-associated proteins. Prolonged exposure to MK801 induces schizophrenia-like behaviors and cognitive deficits in female rats. It also elevates the expression of NLRP3, Caspase-1, IL-1β, and IL-18 proteins in the hippocampus, accompanied by the activation of microglial cells. However, UA treatment can reverse the expression of these inflammatory proteins and the activation of microglial cells induced by MK801. This is the first study to evaluate the effects of UA on behavior and cognition in a female rat model of schizophrenia. The findings indicate that UA mitigates MK-801-induced cognitive deficits in female rats by inhibiting neuroinflammation and microglial activation via modulation of the NLRP3 signaling pathway. These findings offer preclinical data endorsing the possible application of UA as a dietary supplement to prevent cognitive deficits in schizophrenia.
Collapse
Affiliation(s)
- Zhengyuan Huang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan 430060, China; Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Guanghui Chen
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Zhongyu Ren
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Ling Xiao
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan 430060, China; Institute of Neuropsychiatry, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Ziyue Chen
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yinping Xie
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan 430060, China; Institute of Neuropsychiatry, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Gaohua Wang
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan 430060, China; Institute of Neuropsychiatry, Renmin Hospital of Wuhan University, Wuhan 430060, China; Taikang center for life and medical sciences, Wuhan University, Wuhan, 430071, China.
| | - Benhong Zhou
- Department of Psychiatry, Renmin Hospital of Wuhan University, Wuhan 430060, China; Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan 430060, China.
| |
Collapse
|
18
|
Zhang Y, Che N, Wang B, Liu S, Dong Y, Kong X, Yun Y, Li S, Tang XQ, Liao Y. 1,2,4-Trimethoxybenzene ameliorates depression-like behaviors by inhibiting the activation of NLRP3 inflammasome. Int Immunopharmacol 2025; 151:114361. [PMID: 40031427 DOI: 10.1016/j.intimp.2025.114361] [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: 11/21/2024] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 03/05/2025]
Abstract
Our previous works identified that 1,2,4-Trimethoxybenzene (1,2,4-TTB) is an NOD-like receptor (NLR) family pyrin domain-containing-3 (NLRP3) inflammasome-specific inhibitor and ameliorates the severity of experimental autoimmune encephalomyelitis. This study explored whether 1,2,4-TTB has an antidepressive-like role, which depends on suppressing NLRP3 inflammasome. In the current study, the depression-like behaviors induced by lipopolysaccharide (LPS) or fear conditioning were used to determine the antidepressive-like role of 1,2,4-TTB. The results showed that NLRP3 inflammasome is activated in the hippocampus by repetitive injection of LPS. 1,2,4-TTB attenuates the depression-like behaviors and suppresses hippocampal NLRP3 inflammasome activation in LPS-induced depression model mice. In addition, we proved that 1,2,4-TTB enhances the fear memory extinction and alleviates anxiety- and depression-like behavior in mice stimulated by fear conditioning. Consistently,1,2,4-TTB inhibited the activation of hippocampal NLRP3 inflammasome in mice subjected to fear conditioning. Therefore, our study demonstrates that 1,2,4-TTB mitigates depression-like behaviors, depending on its inhibitory role in NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Neurology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, P.R. China.Hunan, Provincial Key Laboratory of Basic and Clinical Pharmacological Research of Gastrointestinal Cancer, The Second Affiliated Hospital, University of South China, Hengyang 421001, Hunan, China; Institute of Neuroscience, Hengyang Medical School, University of South China, 28 W Changsheng Road, Hengyang, 421001, Hunan, People's Republic of China
| | - Na Che
- Institute of Neuroscience, Hengyang Medical School, University of South China, 28 W Changsheng Road, Hengyang, 421001, Hunan, People's Republic of China
| | - Bo Wang
- The First Affiliated Hospital, Institute of Anesthesiology, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, PR China
| | - Shuhan Liu
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, No. 27 Taiping Road, Haidian District, Beijing 100850, China
| | - Yuan Dong
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, No. 27 Taiping Road, Haidian District, Beijing 100850, China
| | - Xiangxi Kong
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, No. 27 Taiping Road, Haidian District, Beijing 100850, China
| | - Yige Yun
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, No. 27 Taiping Road, Haidian District, Beijing 100850, China
| | - Shuoshuo Li
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, No. 27 Taiping Road, Haidian District, Beijing 100850, China.
| | - Xiao-Qing Tang
- Department of Neurology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, P.R. China.Hunan, Provincial Key Laboratory of Basic and Clinical Pharmacological Research of Gastrointestinal Cancer, The Second Affiliated Hospital, University of South China, Hengyang 421001, Hunan, China; Institute of Neuroscience, Hengyang Medical School, University of South China, 28 W Changsheng Road, Hengyang, 421001, Hunan, People's Republic of China.
| | - Yajin Liao
- Department of Neurology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, P.R. China.Hunan, Provincial Key Laboratory of Basic and Clinical Pharmacological Research of Gastrointestinal Cancer, The Second Affiliated Hospital, University of South China, Hengyang 421001, Hunan, China.
| |
Collapse
|
19
|
Ye L, Wang L, Kuang G, Zhang Z, Peng Q, He M, Fan J. IL-27 aggravates acute hepatic injury by promoting macrophage M1 polarization to induce Caspase-11 mediated Pyroptosis in vitro and in vivo. Cytokine 2025; 188:156881. [PMID: 39913960 DOI: 10.1016/j.cyto.2025.156881] [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: 07/24/2024] [Revised: 01/24/2025] [Accepted: 02/01/2025] [Indexed: 03/01/2025]
Abstract
OBJECTIVES Our aim was to explore the IL-27 effect in sepsis (SP)-related acute hepatic injury (AHI) as well as its possible mechanism. MATERIALS AND METHODS Herein, we utilized both wild-type (WT) and IL-27 receptor (WSX-1)-deficient (IL-27R-/-) mice alongside RAW264.7 cells. Our study established an SP-associated AHI model through the intraperitoneal injections of lipopolysaccharide (LPS) + D-galactosamine (D-G). For examining the IL-27 impact on AHI, mice serum and liver tissue samples were gathered. Inflammatory factor levels in the liver and serum were detected using ELISA and immunohistochemistry. Immunofluorescence and Western blot techniques were employed for the detection of protein expression associated with polarization and pyroptosis in the liver, including iNOS, ARG-1, caspase-11, RAGE, and GSDMD. To further verify the IL-27 effects on macrophage polarization and pyroptosis and explore possible mechanisms involved, we used LPS-triggered RAW264.7 macrophages to assess AMPK/SIRT1 expression after IL-27 intervention. This study utilized Compound C (CC) to block the AMPK/SIRT1 pathway. The inflammatory response level and protein expression related to macrophage polarization and pyroptosis were measured again to reveal IL-27 implication in AHI and determine whether its role is associated with the AMPK/SIRT1 pathway. RESULTS The results revealed that IL-27 exacerbated systemic inflammation and liver damage in AHI mice by promoting M1 macrophage polarization, thereby increasing pro-inflammatory phenotype macrophages (M1). This further exacerbated the inflammatory response and pyroptosis in vivo and in vitro. Additionally, IL-27 down-regulated p-AMPK and SIRT1 protein expression while overexpressing macrophage inflammatory mediators including IL-1β/6 and TNFα. Furthermore, IL-27 promoted increased RAGE and caspase-11 protein expression, aggravating macrophage pyroptosis. Employing CC to block the AMPK pathway further aggravated M1 macrophage polarization and pyroptosis in vitro and in vivo, ultimately worsening liver injury. CONCLUSIONS Here, IL-27 aggravates AHI by promoting macrophage M1 polarization to induce caspase-11-mediated pyroptosis in vitro and in vivo, which may be linked to the AMPK/SIRT1 signaling pathway.
Collapse
Affiliation(s)
- Lin Ye
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, No.1, Youyi Road, Yuzhong District, Chongqing City 400016, China; The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical Universit, Chongqing City, China
| | - Liuyang Wang
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, No.1, Youyi Road, Yuzhong District, Chongqing City 400016, China; The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical Universit, Chongqing City, China
| | - Gang Kuang
- The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical Universit, Chongqing City, China; Department of Critical Care Medicine, Affiliated Dazu's Hospital of Chongqing Medical University, No. 1073, The second Ring South Road, Tangxiang Street, DaZu District, Chongqing City 402360, China
| | - Zhijiao Zhang
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, No.1, Youyi Road, Yuzhong District, Chongqing City 400016, China; The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical Universit, Chongqing City, China
| | - Qiaozhi Peng
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, No.1, Youyi Road, Yuzhong District, Chongqing City 400016, China; The Chongqing Key Laboratory of Translational Medicine in Major Metabolic Diseases, The First Affiliated Hospital of Chongqing Medical Universit, Chongqing City, China
| | - Miao He
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Jing Fan
- Department of Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, No.1, Youyi Road, Yuzhong District, Chongqing City 400016, China.
| |
Collapse
|
20
|
Li G, Guan Y, Xu L, Peng G, Han Q, Wang T, Xu Z, Wen X, Lou H, Shen T. Total alkaloids from Thesium chinense inhibit lipopolysaccharide-induced respiratory inflammation by modulating Nrf2/NF-κB/NLRP3 signaling pathway. Chin J Nat Med 2025; 23:421-430. [PMID: 40274345 DOI: 10.1016/s1875-5364(25)60834-x] [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: 01/11/2024] [Revised: 04/07/2024] [Accepted: 04/23/2024] [Indexed: 04/26/2025]
Abstract
Inflammation plays a pivotal role in the etiology and progression of various diseases. In traditional Chinese medicine, the whole plants of Thesium chinense Turcz. and its preparations (e.g. Bairui Granules) have been employed to manage inflammatory conditions. While flavonoids were previously considered the primary anti-inflammatory components, other potentially active constituents have been largely overlooked and not thoroughly investigated. This study presents a novel finding that the total alkaloids of T. chinense (BC-Alk) are potent active substances underlying the traditional and clinical applications of T. chinense and Bairui Granules as anti-inflammatory agents. UPLC-MS/MS analysis identified the composition of BC-Alk as quinolizidine alkaloids. The anti-inflammatory efficacy of BC-Alk was evaluated using a lipopolysaccharide (LPS)-induced lung inflammation model in mice. Results demonstrated that BC-Alk significantly mitigated LPS-induced lung inflammation, attenuated the overproduction of IL-1β and the overproduction of inflammatory factors (TNF-α), and ameliorated lung tissue hyperplasia in mice in vivo. Mechanistic studies in vitro revealed that BC-Alk upregulated the expression of Nrf2 and its downstream proteins NQO1 and glutamate-cystine ligase and modifier subunit (GCLM), inhibited NF-κB phosphorylation, and suppressed NLRP3 activation. Collectively, these findings indicate that BC-Alk exerts potent inhibitory effects against lung inflammation by modulating Nrf2, NF-κB, and NLRP3 pathways. This study provides new insights into the anti-inflammatory constituents of T. chinense and Bairui Granules.
Collapse
Affiliation(s)
- Guohui Li
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; Department of Pharmacy, Jinan Maternity and Child Care Hospital Affiliated to Shandong First Medical University, Jinan 250218, China
| | - Yueqin Guan
- Jiuhua Huayuan Pharmaceutical Co., Ltd., Chuzhou 239011, China
| | - Lintao Xu
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Guangcheng Peng
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Qingtong Han
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Tian Wang
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Zhenpeng Xu
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Xuesen Wen
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Hongxiang Lou
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China.
| | - Tao Shen
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China.
| |
Collapse
|
21
|
Xu J, Wang B, Ao H. Corticosterone effects induced by stress and immunity and inflammation: mechanisms of communication. Front Endocrinol (Lausanne) 2025; 16:1448750. [PMID: 40182637 PMCID: PMC11965140 DOI: 10.3389/fendo.2025.1448750] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 02/28/2025] [Indexed: 04/05/2025] Open
Abstract
The body instinctively responds to external stimuli by increasing energy metabolism and initiating immune responses upon receiving stress signals. Corticosterone (CORT), a glucocorticoid (GC) that regulates secretion along the hypothalamic-pituitary-adrenal (HPA) axis, mediates neurotransmission and humoral regulation. Due to the widespread expression of glucocorticoid receptors (GR), the effects of CORT are almost ubiquitous in various tissue cells. Therefore, on the one hand, CORT is a molecular signal that activates the body's immune system during stress and on the other hand, due to the chemical properties of GCs, the anti-inflammatory properties of CORT act as stabilizers to control the body's response to stress. Inflammation is a manifestation of immune activation. CORT plays dual roles in this process by both promoting inflammation and exerting anti-inflammatory effects in immune regulation. As a stress hormone, CORT levels fluctuate with the degree and duration of stress, determining its effects and the immune changes it induces. The immune system is essential for the body to resist diseases and maintain homeostasis, with immune imbalance being a key factor in the development of various diseases. Therefore, understanding the role of CORT and its mechanisms of action on immunity is crucial. This review addresses this important issue and summarizes the interactions between CORT and the immune system.
Collapse
Affiliation(s)
- Jingyu Xu
- School of Public Health and Management, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Baojuan Wang
- Department of Reproductive Medicine, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Haiqing Ao
- School of Public Health and Management, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
22
|
Beesetti S. Ubiquitin Ligases in Control: Regulating NLRP3 Inflammasome Activation. FRONT BIOSCI-LANDMRK 2025; 30:25970. [PMID: 40152367 DOI: 10.31083/fbl25970] [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: 08/02/2024] [Revised: 09/04/2024] [Accepted: 09/11/2024] [Indexed: 03/29/2025]
Abstract
Ubiquitin ligases play pivotal roles in the regulation of NLR family pyrin domain containing 3 (NLRP3) inflammasome activation, a critical process in innate immunity and inflammatory responses. This review explores the intricate mechanisms by which various E3 ubiquitin ligases exert both positive and negative influences on NLRP3 inflammasome activity through diverse post-translational modifications. Negative regulation of NLRP3 inflammasome assembly is mediated by several E3 ligases, including F-box and leucine-rich repeat protein 2 (FBXL2), tripartite motif-containing protein 31 (TRIM31), and Casitas B-lineage lymphoma b (Cbl-b), which induce K48-linked ubiquitination of NLRP3, targeting it for proteasomal degradation. Membrane-associated RING-CH 7 (MARCH7) similarly promotes K48-linked ubiquitination leading to autophagic degradation, while RING finger protein (RNF125) induces K63-linked ubiquitination to modulate NLRP3 function. Ariadne homolog 2 (ARIH2) targets the nucleotide-binding domain (NBD) domain of NLRP3, inhibiting its activation, and tripartite motif-containing protein (TRIM65) employs dual K48 and K63-linked ubiquitination to suppress inflammasome assembly. Conversely, Pellino2 exemplifies a positive regulator, promoting NLRP3 inflammasome activation through K63-linked ubiquitination. Additionally, ubiquitin ligases influence other components critical for inflammasome function. TNF receptor-associated factor 3 (TRAF3) mediates K63 polyubiquitination of apoptosis-associated speck-like protein containing a CARD (ASC), facilitating its degradation, while E3 ligases regulate caspase-1 activation and DEAH-box helicase 33 (DHX33)-NLRP3 complex formation through specific ubiquitination events. Beyond direct inflammasome regulation, ubiquitin ligases impact broader innate immune signaling pathways, modulating pattern-recognition receptor responses and dendritic cell maturation. Furthermore, they intricately control NOD1/NOD2 signaling through K63-linked polyubiquitination of receptor-interacting protein 2 (RIP2), crucial for nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and mitogen-activated protein kinase (MAPK) activation. Furthermore, we explore how various pathogens, including bacteria, viruses, and parasites, have evolved sophisticated strategies to hijack the host ubiquitination machinery, manipulating NLRP3 inflammasome activation to evade immune responses. This comprehensive analysis provides insights into the molecular mechanisms underlying inflammasome regulation and their implications for inflammatory diseases, offering potential avenues for therapeutic interventions targeting the NLRP3 inflammasome. In conclusion, ubiquitin ligases emerge as key regulators of NLRP3 inflammasome activation, exhibiting a complex array of functions that finely tune immune responses. Understanding these regulatory mechanisms not only sheds light on fundamental aspects of inflammation but also offers potential therapeutic avenues for inflammatory disorders and infectious diseases.
Collapse
Affiliation(s)
- Swarna Beesetti
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| |
Collapse
|
23
|
Yu K, Fu L, Chao Y, Zeng X, Zhang Y, Chen Y, Gao J, Lu B, Zhu H, Gu L, Xiong X, Hu Z, Hong X, Xiao Y. Deep Learning Enhanced Near Infrared-II Imaging and Image-Guided Small Interfering Ribonucleic Acid Therapy of Ischemic Stroke. ACS NANO 2025; 19:10323-10336. [PMID: 40042964 DOI: 10.1021/acsnano.4c18035] [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: 03/19/2025]
Abstract
Small interfering RNA (siRNA) targeting the NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome has emerged as a promising therapeutic strategy to mitigate infarct volume and brain injury following ischemic stroke. However, the clinical translation of siRNA-based therapies is significantly hampered by the formidable blood-brain barrier (BBB), which restricts drug penetration into the central nervous system. To address this challenge, we have developed an innovative long-circulating near-infrared II (NIR-II) nanoparticle platform YWFC NPs, which is meticulously engineered to enhance BBB transcytosis and enable efficient delivery of siRNA targeting NLRP3 (siNLRP3@YWFC NPs) in preclinical models of ischemic stroke. Furthermore, we integrated advanced deep learning neural network algorithms to optimize in vivo NIR-II imaging of the cerebral infarct penumbra, achieving an improved signal-to-background ratio at 72 h poststroke. In vivo studies employing middle cerebral artery occlusion (MCAO) mouse models demonstrated that image-guided therapy with siNLRP3@YWFC NPs, guided by prolonged NIR-II imaging, resulted in significant therapeutic benefits.
Collapse
MESH Headings
- Animals
- Mice
- RNA, Small Interfering/therapeutic use
- RNA, Small Interfering/genetics
- RNA, Small Interfering/chemistry
- RNA, Small Interfering/administration & dosage
- Ischemic Stroke/diagnostic imaging
- Ischemic Stroke/therapy
- Ischemic Stroke/drug therapy
- Deep Learning
- Nanoparticles/chemistry
- NLR Family, Pyrin Domain-Containing 3 Protein/genetics
- NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors
- NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
- Blood-Brain Barrier/metabolism
- Mice, Inbred C57BL
- Male
- Disease Models, Animal
- Infrared Rays
- Infarction, Middle Cerebral Artery
Collapse
Affiliation(s)
- Kai Yu
- Department of Neurosurgery, Central Laboratory, Renmin Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430060, China
| | - Lidan Fu
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yu Chao
- College of Chemistry and Chemical Engineering, Hubei University, Wuhan 430062, China
| | - Xiaodong Zeng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China
| | - Yonggang Zhang
- Department of Neurosurgery, Central Laboratory, Renmin Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Yuanyuan Chen
- Department of Neurosurgery, Central Laboratory, Renmin Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430060, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China
| | - Jialu Gao
- Department of Neurosurgery, Central Laboratory, Renmin Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430060, China
- Shenzhen Institute of Wuhan University, Shenzhen 518057, China
| | - Binchun Lu
- Department of Precision Instrument, Tsinghua University, Beijing 100084, China
| | - Hua Zhu
- Department of Neurosurgery, Central Laboratory, Renmin Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Lijuan Gu
- Department of Neurosurgery, Central Laboratory, Renmin Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Xiaoxing Xiong
- Department of Neurosurgery, Central Laboratory, Renmin Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Zhenhua Hu
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing 100049, China
- National Key Laboratory of Kidney Diseases, Beijing 100853, China
| | - Xuechuan Hong
- Department of Neurosurgery, Central Laboratory, Renmin Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430060, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China
- Shenzhen Institute of Wuhan University, Shenzhen 518057, China
| | - Yuling Xiao
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430060, China
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China
| |
Collapse
|
24
|
Ma C, Zhang L, Huang Q, Deng Q, Huang F, Xu J. Phenethyl isothiocyanate ameliorates liver injuries secondary to inflammatory bowel disease. Food Funct 2025; 16:2589-2597. [PMID: 40047466 DOI: 10.1039/d4fo04931d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2025]
Abstract
Inflammatory bowel disease (IBD) is often accompanied by secondary liver injury which further evolves into various hepatobiliary disorders. The pathogenesis of secondary liver injury involves many different mechanisms including inflammation, pyroptosis, oxidative stress, and heat shock response. Here, we tested the effect of administration of phenethyl isothiocyanate (PEITC) on secondary liver injury in DSS-induced IBD mice. PEITC supplementation reversed liver injury as determined by hepatic injury-related parameters and histopathological examinations. Severe hepatic inflammation with IBD, evidenced by ubiquitously distributed activated macrophages, increased secretion of pro-inflammatory cytokines (TNF-α, IL-1β and IL-6), enhanced expression of inflammation-related proteins (iNOS and COX-2), and augmented activation of the TLR4/NF-κB signaling pathway, was inhibited by PEITC treatment. PEITC also prevented IBD-induced increases in pyroptosis and oxidative stress in the liver. In addition, impairments of hepatic heat shock response elicited by IBD were restored by PEITC treatment. Taken together, these results suggested that PEITC may be effective as a therapeutic reagent to attenuate secondary liver injury caused by IBD.
Collapse
Affiliation(s)
- Congcong Ma
- Department of Nutriology, Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, 2 Xudong Second Road, Wuhan 430062, P.R. China.
- Hubei Key Laboratory of Lipid Chemistry and Nutrition, Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, Wuhan 430062, P.R. China
| | - Li Zhang
- Academy of Nutrition and Health, Hubei Province Key Laboratory of Occupational Hazard, Identification and Control, School of Public Health, Wuhan University of Science and Technology, Wuhan 430065, China
- Department of Neurology, Hubei Provincial Hospital of Integrated Chinese & Western Medicine, No. 11, Lingjiaohu Road, Wuhan 430015, P.R. China
| | - Qingde Huang
- Department of Nutriology, Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, 2 Xudong Second Road, Wuhan 430062, P.R. China.
- Hubei Key Laboratory of Lipid Chemistry and Nutrition, Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, Wuhan 430062, P.R. China
| | - Qianchun Deng
- Department of Nutriology, Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, 2 Xudong Second Road, Wuhan 430062, P.R. China.
- Hubei Key Laboratory of Lipid Chemistry and Nutrition, Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, Wuhan 430062, P.R. China
| | - Fenghong Huang
- Department of Nutriology, Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, 2 Xudong Second Road, Wuhan 430062, P.R. China.
- Hubei Key Laboratory of Lipid Chemistry and Nutrition, Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, Wuhan 430062, P.R. China
| | - Jiqu Xu
- Department of Nutriology, Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, 2 Xudong Second Road, Wuhan 430062, P.R. China.
- Hubei Key Laboratory of Lipid Chemistry and Nutrition, Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, Wuhan 430062, P.R. China
| |
Collapse
|
25
|
Seo HY, Park JY, Lee SH, Lee HW, Han E, Hwang JS, Kim MK, Jang BK. Clusterin deficiency exacerbates cholestatic liver disease through ER stress and NLRP3 inflammasome activation. Cell Biosci 2025; 15:36. [PMID: 40089787 PMCID: PMC11909925 DOI: 10.1186/s13578-025-01376-z] [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: 10/28/2024] [Accepted: 03/04/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND Cholestatic liver disease, characterized by impaired bile flow, leads to the accumulation of harmful metabolites and toxins, resulting in liver damage. Inflammatory cytokines are crucial for the progression of this condition. Clusterin is a glycoprotein with roles in cell death, lipid transport, and cellular protection. We previously demonstrated that clusterin protects against hepatic steatosis and hepatic fibrosis. This study explored the roles of clusterin in cholestatic liver injury induced by a DDC (3,5-diethoxycarbonyl-1,4-dihydrocollidine) diet. METHODS The study evaluated the impact of clusterin on liver injury in C57BL/6 mice and clusterin-knockout (KO) mice fed a DDC diet for 10-20 days. Primary Kupffer cells (KCs) and hepatocytes (HCs) of these mice were analyzed. Techniques such as Sirius red staining, immunohistochemistry, real-time RT-PCR, enzyme-linked immunosorbent assays, and western blotting were performed to assess the effects of clusterin. RESULTS Clusterin expression was upregulated in the cholestatic liver. Clusterin-KO mice exhibited elevated levels of alanine aminotransferase, aspartate aminotransferase, collagen, and αSMA upon DDC diet-induced liver injury. They also had increased levels of markers of endoplasmic reticulum (ER) stress (CHOP, ATF6, and p-eIF2α) and inflammasome activity (NLRP3, ASC, caspase-1, and interleukin 1 beta (IL1β) protein expression, and IL1β and interleukin 18 secretion). Thapsigargin, an ER stress inducer, heightened NLRP3 inflammasome activation in primary KCs and HCs, which was mitigated by overexpression of clusterin. CONCLUSIONS The absence of clusterin exacerbates ER stress and NLRP3 inflammasome activation in mice fed a DDC diet. Conversely, overexpression of clusterin suppresses these stress responses. Thus, clusterin deficiency is associated with an enhanced inflammasome response in the liver that is linked to upregulation of ER stress.
Collapse
Affiliation(s)
- Hye-Young Seo
- Department of Internal Medicine, School of Medicine, Institute for Medical Science, Keimyung University, Daegu, Korea
| | - Ji Yeon Park
- Department of Internal Medicine, School of Medicine, Institute for Medical Science, Keimyung University, Daegu, Korea
| | - So-Hee Lee
- Department of Internal Medicine, School of Medicine, Institute for Medical Science, Keimyung University, Daegu, Korea
| | - Hye Won Lee
- Department of Pathology, Keimyung University School of Medicine, Daegu, Korea
| | - Eugene Han
- Department of Internal Medicine, School of Medicine, Institute for Medical Science, Keimyung University, Daegu, Korea
| | - Jae Seok Hwang
- Department of Internal Medicine, School of Medicine, Institute for Medical Science, Keimyung University, Daegu, Korea
| | - Mi Kyung Kim
- Department of Internal Medicine, School of Medicine, Institute for Medical Science, Keimyung University, Daegu, Korea.
| | - Byoung Kuk Jang
- Department of Internal Medicine, School of Medicine, Institute for Medical Science, Keimyung University, Daegu, Korea.
| |
Collapse
|
26
|
Al-Jawadri AMH, Karami Z, Haririan I, Akrami M, Gholami M. Development of a bio-inspired phagocytic stable nanoghost with anti-inflammatory properties for management of inflammation in ulcerative colitis. J Drug Target 2025:1-15. [PMID: 40022643 DOI: 10.1080/1061186x.2025.2474644] [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: 10/03/2024] [Revised: 02/22/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
BACKGROUND New bio-mimetic approaches are needed to develop effective delivery systems for inflammation regulation in chronic diseases like ulcerative colitis, avoiding fast clearance by immune system. The cell membrane-coated nanoparticle with a therapeutic payload has been considered as a promising delivery system to address the requirement. METHODS Here, Glibenclamide (GLY)-loaded PLGA nanoparticles (NPs) were constructed by a single emulsion procedure and camouflaged by a layer of monocyte membrane using the extrusion technique to fabricate bio-mimetic nanoghosts (NGs), followed by physiochemical and biological characterizations. RESULTS Upon coating the NPs by the membrane, the hydrodynamic size and zeta potential of NGs was changed. The formation of the shell compartment with diameter of about 15.5nm around NP core was confirmed by TEM. The expression levels of NLRP3, IL-1β, IL-18, caspase-1, TNF-α and IL-6 were decreased upon the NGs treatment. The lower cellular internalization of the NGs exhibited potential for improved circulation stability against macrophage phagocytosis. Treatment of acetic acid-induced UC with NGs exhibited healing of the mucosal lining in the colon tissue. CONCLUSION The monocyte membrane-coated NPs with a sulfonylurea derivatives payload can be considered as an excellent biologically inspired candidate for management of inflammatory diseases like UC via inflammation regulation.
Collapse
Affiliation(s)
| | - Zahra Karami
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Ismaeil Haririan
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Akrami
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdi Gholami
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Toxicology and Poisoning Research, Tehran, Iran
| |
Collapse
|
27
|
Wang H, Ma L, Su W, Liu Y, Xie N, Liu J. NLRP3 inflammasome in health and disease (Review). Int J Mol Med 2025; 55:48. [PMID: 39930811 PMCID: PMC11781521 DOI: 10.3892/ijmm.2025.5489] [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: 09/19/2024] [Accepted: 12/16/2024] [Indexed: 02/13/2025] Open
Abstract
Activation of inflammasomes is the activation of inflammation‑related caspase mediated by the assembly signal of multi‑protein complex and the maturity of inflammatory factors, such as IL‑1β and IL‑18. Among them, the Nod‑like receptor family pyrin domain containing 3 (NLRP3) inflammasome is the most thoroughly studied type of inflammatory corpuscle at present, which is involved in the occurrence and development of numerous human diseases. Therefore, targeting the NLRP3 inflammasome has become the focus of drug development for related diseases. In this paper, the research progress of the NLRP3 inflammasome in recent years is summarized, including the activation and regulation of NLRP3 and its association with diseases. A deep understanding of the regulatory mechanism of NLRP3 will be helpful to the discovery of new drug targets and the development of therapeutic drugs.
Collapse
Affiliation(s)
- Haoran Wang
- Department of Orthopaedics, Tongji Hospital Affiliated to Tongji University, Shanghai 200065, P.R. China
| | - Li Ma
- Department of Oncology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210011, P.R. China
| | - Weiran Su
- Department of Internal Medicine, Jiading District Central Hospital, Shanghai 201800, P.R. China
| | - Yangruoyu Liu
- Department of Orthopaedics, Tongji Hospital Affiliated to Tongji University, Shanghai 200065, P.R. China
| | - Ning Xie
- Department of Orthopaedics, Tongji Hospital Affiliated to Tongji University, Shanghai 200065, P.R. China
| | - Jun Liu
- Department of Orthopaedics, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210011, P.R. China
| |
Collapse
|
28
|
Su J, Zhao L, Fu R, Tang Z. Linking Circadian Rhythms to Gut-Brain Axis Lipid Metabolism Associated With Endoplasmic Reticulum Stress in Alzheimer's Disease. CNS Neurosci Ther 2025; 31:e70329. [PMID: 40059063 PMCID: PMC11890981 DOI: 10.1111/cns.70329] [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: 09/13/2024] [Revised: 02/02/2025] [Accepted: 02/26/2025] [Indexed: 05/13/2025] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is characterized by a decline in cognitive, learning, and memory abilities. Neuroinflammation is associated with the spread of tau tangles in the neocortex of AD, leading to cognitive impairment. Therefore, clarifying the pathogenesis of Neuroinflammation and finding effective treatments are the crucial issues for the clinical management of AD. METHOD We systematically review the latest research on the pathogenesis and therapeutic strategies of AD in PubMed, Web of Science, and Elsevier SD. RESULT In this review, the mechanism of the effect of gut-brain axis lipid metabolism mediated by circadian rhythm on AD was discussed, and we also analysed the effects of inflammation and endoplasmic reticulum stress (ERS) induced by lipid abnormalities on intestinal mucosal barrier and neurodegeneration; furthermore, the importance of lipid homeostasis (phospholipids, fatty acids, sterol) in maintaining the functions of endoplasmic reticulum was emphasized. Meanwhile, as lipid composition affects protein conformation, the membrane phospholipids surrounding sarcoplasmic reticulum Ca2+-ATPase (SERCA) that influence SERCA to release Ca2+ mediating inflammation were also reviewed. CONCLUSION We interpreted the mechanism of action between lipid microdomains and ER membrane proteins, reviewed the role of the new pathway of circadian rhythm, lipid metabolism, intestinal mucosa, and brain signaling in the pathogenesis of AD, and proposed strategies to prevent AD by changing the dietary lipid measures.
Collapse
Affiliation(s)
- Jianhui Su
- School of Marine and BioengineeringYancheng Institute of TechnologyYanchengJiangsuChina
| | - Lanyang Zhao
- School of PharmacyNanjing University of Chinese MedicineNanjingChina
| | - Runze Fu
- School of Marine and BioengineeringYancheng Institute of TechnologyYanchengJiangsuChina
| | - Zhe Tang
- School of Chemistry & Chemical EngineeringYancheng Institute of TechnologyYanchengJiangsuChina
| |
Collapse
|
29
|
Yu L, Sun L, Yu T, Guo A, Wu J, Chen J, Wang Q. CPCGI Alleviates Neural Damage by Modulating Microglial Pyroptosis After Traumatic Brain Injury. CNS Neurosci Ther 2025; 31:e70322. [PMID: 40059065 PMCID: PMC11890976 DOI: 10.1111/cns.70322] [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: 11/20/2024] [Revised: 02/15/2025] [Accepted: 02/18/2025] [Indexed: 05/13/2025] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) is a major global cause of mortality and long-term disability, with limited therapeutic options. Microglial pyroptosis, a form of programmed cell death associated with inflammation, has been implicated in exacerbating neuroinflammation and secondary injury following TBI. Compound porcine cerebroside ganglioside injection (CPCGI) has shown anti-inflammatory and antioxidant properties, but its effects on pyroptosis remain unexplored. This study investigates the role of CPCGI in TBI and its underlying mechanisms. METHODS A controlled cortical impact (CCI) model was utilized to establish TBI in vivo, while lipopolysaccharide (LPS) was used in vitro to induce microglial activation that mimicked TBI conditions. The effects of CPCGI on microglial pyroptosis and inflammatory cytokines were analyzed through immunofluorescence, flow cytometry, western blotting, and quantitative real-time PCR (qRT-PCR). The involvement of the NLRP3 inflammasome in CPCGI's mechanism was examined using NLRP3 overexpression or the NLRP3 agonist BMS-986299. A microglia-neuron interaction model was created, and neuronal injury was assessed with the Cell Counting Kit-8 and Fluoro-Jade C (FJC). RESULTS Treatment with CPCGI resulted in significant improvement in the neurobehavioral outcomes, reduced lesion volume, and decreased neuronal loss following TBI. Notably, TBI induced microglial pyroptosis and the release of pro-inflammatory cytokines, while CPCGI inhibited microglial pyroptosis, thereby mitigating the inflammatory response and reducing neuronal damage. Mechanistically, overexpression of NLRP3 in microglial cells reversed the inhibitory effects of CPCGI on microglial pyroptosis, indicating that CPCGI's inhibition of microglial pyroptosis may be mediated by the NLRP3 inflammasome. Furthermore, NLRP3 overexpression or administration of the NLRP3 agonist BMS-986299 negated the neuroprotective effects of CPCGI in vivo and in vitro. CONCLUSION These findings suggest that CPCGI provides neuroprotection in TBI by targeting NLRP3 inflammasome-mediated microglial pyroptosis, thereby improving the neuroinflammatory microenvironment and promoting neurological recovery. This underscores its potential as a promising candidate for TBI treatment.
Collapse
Affiliation(s)
- Lu‐Lu Yu
- Department of NeurologyBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Lei Sun
- Department of NeurologyZhengzhou University People's HospitalZhengzhouChina
| | - Ting‐Ting Yu
- Department of NeurologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| | - An‐Chen Guo
- China National Clinical Research Center for Neurological DiseasesBeijingChina
- Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain DisordersCapital Medical UniversityBeijingChina
- Beijing Key Laboratory of Drug and Device Research and Development for Cerebrovascular DiseasesBeijingChina
| | - Jian‐Ping Wu
- Department of NeurologyBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
- Beijing Key Laboratory of Drug and Device Research and Development for Cerebrovascular DiseasesBeijingChina
- Advanced Innovation Center for Human Brain ProtectionCapital Medical UniversityBeijingChina
| | - Jun‐Min Chen
- Department of NeurologyBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Qun Wang
- Department of NeurologyBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- China National Clinical Research Center for Neurological DiseasesBeijingChina
- Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain DisordersCapital Medical UniversityBeijingChina
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| |
Collapse
|
30
|
Shi S, Jiang H, Ma W, Guan Z, Han M, Man S, Wu Z, He S. Preclinical studies of natural flavonoids in inflammatory bowel disease based on macrophages: a systematic review with meta-analysis and network pharmacology. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:2293-2318. [PMID: 39422746 DOI: 10.1007/s00210-024-03501-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 09/30/2024] [Indexed: 10/19/2024]
Abstract
Flavonoid is a category of bioactive polyphenolic compounds that are extensively distributed in plants with specific pharmacological properties, such as anti-inflammatory and anti-oxidant. Importantly, natural flavonoids have shown the protected function on the dextran sulfate sodium (DSS)-induced colitis in animals and lipopolysaccharides (LPS)-induced inflammatory response in macrophages. The purpose of this systematic review is to explore the efficacy of natural flavonoids in animal models of IBD (inflammatory bowel disease) and potential mechanisms in macrophages by meta-analysis and network pharmacology in preclinical studies. Relevant foundation studies were searched from January 2010 to November 2023 in databases like PubMed, Elsevier ScienceDirect, and Web of Science. Then, OriginPro software was used to extract values from images, and the analysis was performed using Review Manager 5.3. The retrieved data was analyzed according to the fixed-effects model and random-effects model. Subsequently, heterogeneity was evaluated using the I2 statistics. Lastly, network pharmacology was applied to confirm mechanisms of natural flavonoids on IBD. According to the results of meta-analysis, we found the natural flavonoids exhibited powerful therapeutic effects against IBD, which not only reversed colonic shortness (WMD = 1.33, 95% CI (1.07, 1.59), P < 0.00001), but also reduced histological score (SMD = - 2.66, 95% CI (- 3.77, - 1.95), P < 0.00001) between natural flavonoid treatment groups compared with the experimental IBD model. Furthermore, treatment with natural flavonoids decreased the levels of tumor necrosis factor-α (TNF-α) in macrophages. Mechanistically, our summarized data substantiate that natural flavonoids alleviate LPS-induced M1 macrophage polarization, anti-oxidant, anti-inflammatory, maintain intestinal barrier, and inhibit the activation of the NLR family pyrin domain containing 3 (NLRP3) inflammasome in macrophages. Moreover, the results of network pharmacology also support this. This systematic review demonstrated the efficiency of natural flavonoids in treating IBD in preclinical research by meta-analysis and network pharmacology, which offered supporting evidence for clinical trial implementation. However, some limitations remain present, such as technique quality shortage, missed reports on account of negative results, failure to count sample size, and the risk of bias.
Collapse
Affiliation(s)
- Shasha Shi
- Pharmacy School, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Hao Jiang
- Pharmacy School, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Wenke Ma
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Zitong Guan
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Mengxue Han
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Shuai Man
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
- Key Laboratory of Traditional Chinese Medicine for Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Zhuzhu Wu
- Key Laboratory of Traditional Chinese Medicine for Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Shan He
- Pharmacy School, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| |
Collapse
|
31
|
Balde A, Benjakul S, Nazeer RA. A review on NLRP3 inflammasome modulation by animal venom proteins/peptides: mechanisms and therapeutic insights. Inflammopharmacology 2025; 33:1013-1031. [PMID: 39934538 DOI: 10.1007/s10787-025-01656-7] [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: 12/06/2024] [Accepted: 01/07/2025] [Indexed: 02/13/2025]
Abstract
The venom peptides from terrestrial as well as aquatic species have demonstrated potential in regulating the NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome, a sophisticated assemblage present in immune cells responsible for detecting and responding to external mediators. The NLRP3 inflammasome plays a role in several pathological conditions such as type 2 diabetes, hyperglycemia, Alzheimer's disease, obesity, autoimmune disorders, and cardiovascular disorders. Venom peptides derived from animal venoms have been discovered to selectively induce certain signalling pathways, such as the NLRP3 inflammasome, mitogen-activated protein kinase (MAPK), and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). Experimental evidence has demonstrated that venom peptides can regulate the expression and activation of the NLRP3 inflammasome, resulting in the secretion of pro-inflammatory cytokines including interleukin (IL)-1β and IL-18. Furthermore, these peptides have been discovered to impede the activation of the NLRP3 inflammasome, therefore diminishing inflammation and tissue injury. The functional properties of venom proteins and peptides obtained from snakes, bees, wasps, and scorpions have been thoroughly investigated, specifically targeting the NLRP3 inflammasome pathway, venom proteins and peptides have shown promise as therapeutic agents for the treatment of certain inflammatory disorders. This review discusses the pathophysiology of NLRP3 inflammasome in the onset of various diseases, role of venom as therapeutics. Further, various venom components and their role in the modulation of NLRP3 inflammasome are discoursed. A substantial number of venomous animals and their toxins are yet unexplored, and to comprehensively grasp the mechanisms of action of them and their potential as therapeutic agents, additional research is required which can lead to the development of novel therapeutics.
Collapse
Affiliation(s)
- Akshad Balde
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu, 603 203, India
| | - Soottawat Benjakul
- International Center of Excellence in Seafood Science and Innovation, Faculty of Agro Industry, Prince of Songkla University, Hat Yai, 90110, Songkhla, Thailand
- Department of Food and Nutrition, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Rasool Abdul Nazeer
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu, 603 203, India.
| |
Collapse
|
32
|
Guan Q, Xiong H, Song X, Liu S, Guang Y, Nie Q, Xie Y, Zhang XL. Suppression of NLRP3 inflammasome by a small molecule targeting CK1α-β-catenin-NF-κB and CK1α-NRF2-mitochondrial OXPHOS pathways during mycobacterial infection. Front Immunol 2025; 16:1553093. [PMID: 40092991 PMCID: PMC11906677 DOI: 10.3389/fimmu.2025.1553093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 02/10/2025] [Indexed: 03/19/2025] Open
Abstract
Introduction Pyroptosis is an important inflammatory form of cell death and Mycobacterium tuberculosis (M.tb) chronic infection triggers excessive inflammatory pyroptosis of macrophages. Our previous research has confirmed that a small compound pyrvinium pamoate (PP) could inhibit inflammatory pathological changes and mycobacterial burden in M.tb-infected mice, but the potential mechanism of PP for inhibiting M.tb-induced inflammation remains unexplored. Methods The effects of PP on the NLRP3-ASC-Casp1 inflammasome assembly and activation, gasdermin D (GSDMD) mediated pyroptosis and inflammatory cytokines expression were assessed in human THP-1-derived macrophages after M.tb H37Rv/H37Ra/ Salmonella typhimurium (S. typhimurium) infection or LPS treatment by Transcriptome sequencing, RT-qPCR, Co-immunoprecipitation and Western Blot (WB) analysis. The lactate dehydrogenase (LDH) release assay was used to evaluate the CC50 of PP in M.tb-infected THP-1 cells. Results We found that M.tb/S. typhimurium infection and LPS treatment significantly activate NLRP3-ASC-Casp1 inflammasome activation, GSDMD-mediated pyroptosis and inflammatory cytokines (IL-1β and IL-18) expression in macrophages, whereas PP could suppress these inflammatory effects in a dose dependent manner. Regarding the PP-inhibition mechanism, we further found that this inhibitory activity is mediated through the PP-targeting casein kinase 1A1 (CK1α)-β-catenin-NF-κB pathway and CK1α-NRF2-mitochondrial oxidative phosphorylation (OXPHOS) pathway. In addition, a CK1α specific inhibitor D4476 or CK1α siRNA could reverse these inhibitory effects of PP on bacteria-induced inflammatory responses in macrophages. Conclusions This study reveals a previously unreported mechanism that pyrvinium can inhibit NLRP3 inflammasome and GSDMD-IL-1β inflammatory pyroptosis via targeting suppressing CK1α-β-catenin-NF-κB and CK1α-NRF2-mitochondrial OXPHOS pathways, suggesting that pyrvinium pamoate holds great promise as a host directed therapy (HDT) drug for mycobacterial-induced excessive inflammatory response.
Collapse
Affiliation(s)
- Qing Guan
- State Key Laboratory of Virology and Biosafety, Hubei Province Key Laboratory of Allergy and Immunology, Department of Immunology Wuhan University Taikang Medical School (School of Basic Medical Sciences) Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Department of Allergy Zhongnan Hospital, Wuhan University School of Medicine, Wuhan, China
| | - Huan Xiong
- State Key Laboratory of Virology and Biosafety, Hubei Province Key Laboratory of Allergy and Immunology, Department of Immunology Wuhan University Taikang Medical School (School of Basic Medical Sciences) Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Department of Allergy Zhongnan Hospital, Wuhan University School of Medicine, Wuhan, China
| | - Xiangyu Song
- State Key Laboratory of Virology and Biosafety, Hubei Province Key Laboratory of Allergy and Immunology, Department of Immunology Wuhan University Taikang Medical School (School of Basic Medical Sciences) Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Department of Allergy Zhongnan Hospital, Wuhan University School of Medicine, Wuhan, China
| | - Sheng Liu
- State Key Laboratory of Virology and Biosafety, Hubei Province Key Laboratory of Allergy and Immunology, Department of Immunology Wuhan University Taikang Medical School (School of Basic Medical Sciences) Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Department of Allergy Zhongnan Hospital, Wuhan University School of Medicine, Wuhan, China
- Department of Laboratory Medicine, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yuanjun Guang
- State Key Laboratory of Virology and Biosafety, Hubei Province Key Laboratory of Allergy and Immunology, Department of Immunology Wuhan University Taikang Medical School (School of Basic Medical Sciences) Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Department of Allergy Zhongnan Hospital, Wuhan University School of Medicine, Wuhan, China
| | - Qi Nie
- Department of Multidrug-Resistant and Rifampicin-Resistant Tuberculosis (MDR/RR-TB), Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, Hubei, China
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Yan Xie
- State Key Laboratory of Virology and Biosafety, Hubei Province Key Laboratory of Allergy and Immunology, Department of Immunology Wuhan University Taikang Medical School (School of Basic Medical Sciences) Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Department of Allergy Zhongnan Hospital, Wuhan University School of Medicine, Wuhan, China
| | - Xiao-Lian Zhang
- State Key Laboratory of Virology and Biosafety, Hubei Province Key Laboratory of Allergy and Immunology, Department of Immunology Wuhan University Taikang Medical School (School of Basic Medical Sciences) Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Department of Allergy Zhongnan Hospital, Wuhan University School of Medicine, Wuhan, China
| |
Collapse
|
33
|
Ou H, Wu Z, Ning J, Huang Q, Wang W, Yang G, Zhou Y, Hou A, Li P, Chen L, Jin WB. In vitro and in vivo characterization of oridonin analogs as anti-inflammatory agents that regulate the NF-κB and NLRP3 inflammasome axis. Front Pharmacol 2025; 16:1512740. [PMID: 40083382 PMCID: PMC11903421 DOI: 10.3389/fphar.2025.1512740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 01/28/2025] [Indexed: 03/16/2025] Open
Abstract
Introduction A series of oridonin hybrids were synthesized and evaluated for anti-inflammatory potential, focusing on their ability to inhibit NO production in RAW264.7 cells and their therapeutic prospects for NLRP3-driven disorders. Methods Anti-inflammatory activity was assessed by measuring NO inhibition in LPS-stimulated RAW264.7 cells. The most active compound, 4c, was further analyzed using ELISA and WB to evaluate its effects on inflammatory proteins (p-NF-κB, p-IκB, NLRP3, IL-6, IL-1β, COX-2, iNOS). In vivo efficacy was tested in a murine acute lung injury model, with RT‒qPCR and WB used to assess inflammatory markers in lung tissues. Molecular docking predicted 4c's binding mode with NLRP3, while RNA-seq and RT‒qPCR identified differentially expressed genes. Results Compound 4c significantly inhibited NO production and suppressed key inflammatory proteins in vitro. In vivo, it alleviated acute lung injury, reduced IL-6 and TNF-α mRNA levels, and inhibited NLRP3, p-NF-κB, and IL-6 protein expression. Docking suggested covalent binding to NLRP3. RNA-seq revealed 4c upregulated Trdc, Stfa2, and Gsta2 while downregulating Spib, Csf2, and Nr4a1. Discussion Compound 4c demonstrates potent anti-inflammatory effects via NLRP3 pathway inhibition and modulation of inflammatory genes. These findings highlight oridonin hybrids, particularly 4c, as promising candidates for NLRP3-driven inflammatory disorders, warranting further investigation.
Collapse
Affiliation(s)
- Huiping Ou
- Faculty of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
- School of Food and Drug, Shenzhen Polytechnic University, Shenzhen, Guangdong, China
| | - Zhanpan Wu
- Faculty of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Jinhua Ning
- Faculty of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Qiufeng Huang
- School of Food and Drug, Shenzhen Polytechnic University, Shenzhen, Guangdong, China
| | - Wancun Wang
- Faculty of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Guochun Yang
- Faculty of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Yingxun Zhou
- Faculty of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Anguo Hou
- Faculty of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Peng Li
- School of Food and Drug, Shenzhen Polytechnic University, Shenzhen, Guangdong, China
| | - Lingyun Chen
- Faculty of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Wen Bin Jin
- Faculty of Chinese Materia Medica, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| |
Collapse
|
34
|
He KL, Yu X, Xia L, Xie YD, Qi EB, Wan L, Hua XM, Jing CH. A new perspective on the regulation of neuroinflammation in intracerebral hemorrhage: mechanisms of NLRP3 inflammasome activation and therapeutic strategies. Front Immunol 2025; 16:1526786. [PMID: 40083546 PMCID: PMC11903264 DOI: 10.3389/fimmu.2025.1526786] [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: 11/12/2024] [Accepted: 02/10/2025] [Indexed: 03/16/2025] Open
Abstract
Intracerebral hemorrhage (ICH), a specific subtype within the spectrum of stroke disorders, is characterized by its high mortality and significant risk of long-term disability. The initiation and progression of neuroinflammation play a central and critical role in the pathophysiology of ICH. The NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome, a protein complex involved in initiating inflammation, is the central focus of this article. Microglia and astrocytes play critical roles in the inflammatory damage process associated with neuroinflammation. The NLRP3 inflammasome is expressed within both types of glial cells, and its activation drives these cells toward a pro-inflammatory phenotype, which exacerbates inflammatory damage in the brain. However, the regulatory relationship between these two cell types remains to be explored. Targeting NLRP3 inflammasomes in microglia or astrocytes may provide an effective approach to mitigate neuroinflammation following ICH. This article first provides an overview of the composition and activation mechanisms of the NLRP3 inflammasome. Subsequently, it summarizes recent research findings on novel signaling pathways that regulate NLRP3 inflammasome activity. Finally, we reviewed recent progress in NLRP3 inflammasome inhibitors, highlighting the clinical translation potential of certain candidates. These inhibitors hold promise as innovative strategies for managing inflammation following ICH.
Collapse
Affiliation(s)
- Kai-long He
- Department of Neurosurgery, XinHua Hospital, Affiliated to Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Xian Yu
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Lei Xia
- Department of Neurosurgery, XinHua Hospital, Affiliated to Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Yan-dong Xie
- Department of Neurosurgery, XinHua Hospital, Affiliated to Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - En-bo Qi
- Department of Neurosurgery, XinHua Hospital, Affiliated to Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Liang Wan
- Department of Neurosurgery, XinHua Hospital, Affiliated to Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Xu-ming Hua
- Department of Neurosurgery, XinHua Hospital, Affiliated to Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Chao-hui Jing
- Department of Neurosurgery, XinHua Hospital, Affiliated to Shanghai JiaoTong University School of Medicine, Shanghai, China
| |
Collapse
|
35
|
Wang F, Liu Y, Zhao Y, Zheng H, Zhang L. The absence of IRG1 exacerbates bone loss in a mouse model of ovariectomy-induced osteoporosis by increasing osteoclastogenesis through the potentiation of NLRP3 inflammasome activation. Int Immunopharmacol 2025; 148:114099. [PMID: 39870006 DOI: 10.1016/j.intimp.2025.114099] [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: 09/25/2024] [Revised: 01/13/2025] [Accepted: 01/14/2025] [Indexed: 01/29/2025]
Abstract
The immune-responsive gene 1 (IRG1) protein plays a role in various pathological processes by connecting cellular metabolism to a range of cellular activities through the production of itaconate. Recent studies have highlighted the significance of IRG1 and itaconate in bone metabolism and homeostasis. However, the precise role of IRG1 in osteoporosis remains inadequately documented. This study aimed to determine the role of IRG1 in osteoporosis through the utilization of IRG1 knockout (KO) mice and a model of ovariectomy (OVX)-induced osteoporosis. The expression of IRG1 was found to be higher in the bone tissues of postmenopausal osteoporotic mice induced by OVX in comparison to sham control mice. When compared to wild type (WT) mice, OVX-induced bone loss was significantly worse in IRG1 KO mice, and this was accompanied by an increase in osteoclastogenesis and bone resorption. However, the loss of bone and the process of osteoclastogenesis and bone resorption were effectively reversed when the IRG1 KO mice were replenished with itaconate. The osteoclastogenesis induced by receptor activator of nuclear factor kappa-Β ligand (RANKL) in bone marrow-derived macrophages (BMMs) was found to be enhanced by IRG1 deficiency, which could be reversed through the replenishment of itaconate. Further investigation revealed that IRG1 deficiency potentiated the activation of NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome. The inhibition of NLRP3 inflammasome using a targeted inhibitor significantly ameliorated RANKL-induced osteoclastogenesis in IRG1 KO BMMs. Overall, this study highlights the significance of IRG1 in regulating osteoclastogenesis and proposes it as a potential target for osteoporosis treatment.
Collapse
Affiliation(s)
- Fang Wang
- Department of Endocrinology, Sichuan Provincial People's Hospital, Southwest Medical University, Luzhou, Sichuan 646099, China
| | - Yanqiao Liu
- Department of Endocrinology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Yanqin Zhao
- Department of Endocrinology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Hongyin Zheng
- Department of Nuclear Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Lei Zhang
- Department of Endocrinology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China.
| |
Collapse
|
36
|
Chen K, Ying J, Zhu J, Chen L, Liu R, Jing M, Wang Y, Zhou K, Wu L, Wu C, Xiao J, Ni W. Urolithin A alleviates NLRP3 inflammasome activation and pyroptosis by promoting microglial mitophagy following spinal cord injury. Int Immunopharmacol 2025; 148:114057. [PMID: 39827665 DOI: 10.1016/j.intimp.2025.114057] [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: 01/02/2025] [Accepted: 01/06/2025] [Indexed: 01/22/2025]
Abstract
Spinal cord injury (SCI) is a potentially fatal condition that often results in loss of motor and sensory functions, thereby significantly burdening global health initiatives. Urolithin A (UA), an intestinal microbial metabolite of ellagic acid, is known for its potent anti-inflammatory properties in chronic inflammation contexts. UA treatment in humans induces a molecular signature of improved mitochondrial and cellular health. Yet, its effects on acute inflammation following SCI remain unclear. In this study, we developed an impact-induced mouse model for SCI and treated the injured mice with UA (50 mg/kg/d, till 8 weeks) via intragastric administration. Furthermore, we subjected BV2 cells to lipopolysaccharide and adenosine 5'-triphosphate to simulate the post-injury inflammatory response. Our results demonstrated that pre-treatment with UA (10 μM) effectively inhibited NLRP3 inflammasome activation in LPS-primed BV2 cells. This inhibition was evidenced by reduced cleaved Caspase-1 and mature IL-1β release, diminished ASC speck formation, and decreased gasdermin D (GSDMD)-mediated pyroptosis. Additionally, UA treatment restored mitochondrial activity and ROS production attenuated by NLRP3 activation, increased LC3-II expression, and enhanced LC3 co-localization with mitochondria. 3-Methyladenine (3-MA), an autophagy inhibitor, can partially reverse the stimulatory effect of UA on mitophagy, as well as the inhibitory effect of UA on pyroptosis. This study highlighted the protective role of UA against SCI through its promotion of mitophagy, which in turn inhibits NLRP3 inflammasome activation and pyroptosis.
Collapse
Affiliation(s)
- Kongbin Chen
- Department of Orthopedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000 China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000 China
| | - Jiahao Ying
- Department of Orthopedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000 China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000 China
| | - Jiangwei Zhu
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000 China
| | - Liang Chen
- Department of Orthopedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000 China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000 China
| | - Rongjie Liu
- Department of Orthopedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000 China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000 China
| | - Mengqi Jing
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000 China
| | - Yuchao Wang
- Department of Orthopedic, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116600, China
| | - Kailiang Zhou
- Department of Orthopedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000 China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000 China
| | - Long Wu
- Department of Orthopedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000 China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000 China.
| | - Chenyu Wu
- Department of Orthopedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000 China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000 China; Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000 China.
| | - Jian Xiao
- Department of Orthopedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000 China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000 China; Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000 China.
| | - Wenfei Ni
- Department of Orthopedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000 China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000 China; Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000 China.
| |
Collapse
|
37
|
Lian Y, Zhang H, Xing W, Li S, Lai X, Jia S, Shang J, Liu H. Global Research Trends and Focus on the Link Between Heart Failure and NLRP3 Inflammasome: A Bibliometric Analysis From 2010 to 2024. J Multidiscip Healthc 2025; 18:697-710. [PMID: 39949850 PMCID: PMC11822290 DOI: 10.2147/jmdh.s505356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
Background Heart failure (HF) is characterized by elevated morbidity, mortality, and rehospitalization frequencies. This condition imposes a considerable medical burden and fiscal strain on society. Inflammation plays a crucial role in the inception, advancement, and outcome of HF. Despite mounting evidence demonstrating the pivotal function of the NLRP3 inflammasome in HF, a thorough bibliometric examination of research focal points and trajectories in this domain has yet to be undertaken. Methods Publications related to the NLRP3 inflammasome in HF were retrieved from the Web of Science database spanning 2010-2024. The acquired data were subsequently analyzed utilizing various visualization instruments, including Citespace, VOSviewer, Scimago Graphica, and Microsoft Office Excel 2021. Results A total of 282 papers were included in the analysis, authored by 2,130 researchers from 500 institutions across 34 nations/regions. China emerged as a significant contributor to this field, producing the highest number of outputs. Antonio Abbate was identified as the most prolific author. Virginia Commonwealth University and Wuhan University were the institutions with the highest publication output. INTERNATIONAL IMMUNOPHARMACOLOGY was the periodical with the most numerous publications in this sphere. CIRCULATION, however, received the highest number of citations, indicating its substantial influence on investigations in this field. Contemporary research focal points primarily concentrate on the activation and inhibition pathways of the NLRP3 inflammasome, the exploration of novel HF targets, and the association between HF and mitochondrial function. Future research trajectories are likely to encompass investigations into the relationship between HF and pyroptosis, as well as clinical studies on pharmaceuticals targeting the NLRP3 inflammasome as a therapeutic approach for HF. Conclusion This investigation provides a comprehensive bibliometric analysis and synopsis of NLRP3 inflammable-related studies in HF. The findings offer a conceptual foundation for further research on the NLRP3 inflammasome in HF and provide valuable guidance for future research directions in this domain.
Collapse
Affiliation(s)
- YanJie Lian
- Department of Cardiovascular, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, People’s Republic of China
- Graduate School of Capital Medical University, Beijing, 100069, People’s Republic of China
| | - Heyi Zhang
- Department of Cardiovascular, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, People’s Republic of China
- Graduate School of Capital Medical University, Beijing, 100069, People’s Republic of China
| | - Wenlong Xing
- Department of Cardiovascular, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, People’s Republic of China
| | - Sinai Li
- Beijing Institute of Traditional Chinese Medicine, Beijing, 100010, People’s Republic of China
| | - Xiaolei Lai
- Department of Cardiovascular, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, People’s Republic of China
| | - Sihan Jia
- Department of Cardiovascular, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, People’s Republic of China
| | - JuJu Shang
- Department of Cardiovascular, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, People’s Republic of China
| | - Hongxu Liu
- Department of Cardiovascular, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, People’s Republic of China
| |
Collapse
|
38
|
Anjum I, Nasir A, Naseer F, Ibrahim A, Rehman B, Bashir F, Tul Ain Q. Exploring the anti-inflammatory effects of phytochemicals in attenuating interstitial cystitis-a literature review. Front Pharmacol 2025; 16:1483548. [PMID: 39974737 PMCID: PMC11836544 DOI: 10.3389/fphar.2025.1483548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 01/07/2025] [Indexed: 02/21/2025] Open
Abstract
Interstitial cystitis is a fierce syndrome affecting the quality of life of thousands of individuals around the globe. It causes immense pain in the bladder and associated viscera along with inflammation-like lesions. The current medicinal and pharmacological research focuses on the protective and curative effects of phytochemicals in several ailments. Phytochemicals derived from many medicinal plants have shown potent outcomes in protection against various pathological conditions including interstitial cystitis. This review has summarized the insights of in vitro and in vivo studies regarding the effects of phytochemicals in fading the inflammation in bladder tissue and exhibiting a protective effect on the urothelium. Hemorrhagic cystitis is a common manifestation in patients undergoing chemotherapy with cyclophosphamide and related alkylating agents. Sodium 2-mercaptoethane sulfonate (Mesna) has traditionally been employed in clinical practice to counter cyclophosphamide-induced cystitis in humans. However, cyclophosphamide has been employed in developing animal models of interstitial cystitis in in vivo studies. Phytochemicals including quercetin, beta-caryophyllene, curcumol, boswellic acid, caftaric acid, some flavonoids and other secondary metabolites being a consequential component of numerous medicinal plants, have displayed a significant reduction in the levels of proinflammatory cytokines including TNF-α, NFĸB, IL-1β, NLRP3 inflammasome, IL-6, IL-2, matrix metalloproteinases etc. Uroprotective outcomes of these phytochemicals have been found to result in diminished oxidative stress and restoration of glutathione, superoxide dismutase, and related proteins in the inflamed bladder tissue. Many in vivo studies involving cyclophosphamide-induced interstitial cystitis have confirmed these findings. The coupling of phytotherapy with novel drug delivery systems such as nanoparticles, liposomes, nanotubes, quantum dots, etc. can help translate these beneficial effects of phytochemicals into clinical practice. Further investigations of these phytochemicals can provide intuition regarding the development of newer drug molecules having exclusive activity for attenuating interstitial cystitis.
Collapse
Affiliation(s)
- Irfan Anjum
- Department of Basic Medical Sciences, Shifa College of Pharmaceutical Sciences, Shifa Tameer-e-Millat University, Islamabad, Pakistan
| | - Ayesha Nasir
- Department of Basic Medical Sciences, Shifa College of Pharmaceutical Sciences, Shifa Tameer-e-Millat University, Islamabad, Pakistan
| | - Faiza Naseer
- Department of Basic Medical Sciences, Shifa College of Pharmaceutical Sciences, Shifa Tameer-e-Millat University, Islamabad, Pakistan
- Department of Biosciences, Shifa Tameer e Millat University, Islamabad, Pakistan
| | - Ahsan Ibrahim
- Department of Basic Medical Sciences, Shifa College of Pharmaceutical Sciences, Shifa Tameer-e-Millat University, Islamabad, Pakistan
| | - Bisma Rehman
- Department of Basic Medical Sciences, Shifa College of Pharmaceutical Sciences, Shifa Tameer-e-Millat University, Islamabad, Pakistan
| | - Fawad Bashir
- Department of Basic Medical Sciences, Shifa College of Pharmaceutical Sciences, Shifa Tameer-e-Millat University, Islamabad, Pakistan
| | - Qura Tul Ain
- Shifa College of Medicine, Shifa Tameer-e-Millat University, Islamabad, Pakistan
| |
Collapse
|
39
|
Wang S, Huang S, Peng F, Wu Y, Pan W, Huang Y, Luo P. Design, Synthesis and Biological Activities Evaluation of Novel Pterostilbene-Urea Derivatives as Potential Anti-Inflammatory Agents. Chem Biodivers 2025; 22:e202402016. [PMID: 39392379 DOI: 10.1002/cbdv.202402016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 10/07/2024] [Accepted: 10/10/2024] [Indexed: 10/12/2024]
Abstract
1. The toxicity of derivatives was removed by the reasonable modification of bioactive skeleton. 2. As potential COX-2 inhibitor with IC50 values ranging from 39.42 to 179.84 nM/L, compounds (Q4-Q10, Q20) exhibited superior anti-inflammatory activity at low micromolar concentrations. 3. Q7 (IC50 (COX-2)= 61.05 nM/L), Q10 (IC50 (COX-2)= 54.68 nM/L) and Q20 (IC50 (COX-2)= 39.42 nM/L) showed stronger COX-2 inhibitory abilities than Celecoxib (IC50 (COX-2)= 67.89 nM/L). 4. The strongest anti-inflammatory agent, Q20 (IC50 NO= 9.96 μM/L, IC50 (COX-2)= 39.42 nM/L) effectively inhibited the secretion of IL-1β and TNF-α, exhibited the IC50 values of 12.30 and 9.07 μM/L respectively. 5. Q20 exerted as anti-inflammatory actives via targeting COX-2, down-regulating iNOS and TLR4 protein, and inhibiting the activation of NLRP3 inflammasome and NF-κB signal pathway.
Collapse
Affiliation(s)
- Shouchuan Wang
- Zhuang Medical College, Guangxi University of Chinese Medicine, Nanning, 530001, China
| | - Shaoling Huang
- Zhuang Medical College, Guangxi University of Chinese Medicine, Nanning, 530001, China
| | - Feng Peng
- Zhuang Medical College, Guangxi University of Chinese Medicine, Nanning, 530001, China
| | - Yanchun Wu
- Zhuang Medical College, Guangxi University of Chinese Medicine, Nanning, 530001, China
| | - Weigao Pan
- Zhuang Medical College, Guangxi University of Chinese Medicine, Nanning, 530001, China
| | - Yunhou Huang
- Zhuang Medical College, Guangxi University of Chinese Medicine, Nanning, 530001, China
| | - Peng Luo
- Zhuang Medical College, Guangxi University of Chinese Medicine, Nanning, 530001, China
| |
Collapse
|
40
|
Qian W, Zhang B, Gao M, Wang Y, Shen J, Liang D, Wang C, Wei W, Pan X, Yan Q, Sun D, Zhu D, Cheng H. Supramolecular prodrug inspiried by the Rhizoma Coptidis - Fructus Mume herbal pair alleviated inflammatory diseases by inhibiting pyroptosis. J Pharm Anal 2025; 15:101056. [PMID: 39974618 PMCID: PMC11835567 DOI: 10.1016/j.jpha.2024.101056] [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: 03/25/2024] [Revised: 07/25/2024] [Accepted: 07/28/2024] [Indexed: 02/21/2025] Open
Abstract
Sustained inflammatory responses are closely related to various severe diseases, and inhibiting the excessive activation of inflammasomes and pyroptosis has significant implications for clinical treatment. Natural products have garnered considerable concern for the treatment of inflammation. Huanglian-Wumei decoction (HLWMD) is a classic prescription used for treating inflammatory diseases, but the necessity of their combination and the exact underlying anti-inflammatory mechanism have not yet been elucidated. Inspired by the supramolecular self-assembly strategy and natural drug compatibility theory, we successfully obtained berberine (BBR)-chlorogenic acid (CGA) supramolecular (BCS), which is an herbal pair from HLWMD. Using a series of characterization methods, we confirmed the self-assembly mechanism of BCS. BBR and CGA were self-assembled and stacked into amphiphilic spherical supramolecules in a 2:1 molar ratio, driven by electrostatic interactions, hydrophobic interactions, and π-π stacking; the hydrophilic fragments of CGA were outside, and the hydrophobic fragments of BBR were inside. This stacking pattern significantly improved the anti-inflammatory performance of BCS compared with that of single free molecules. Compared with free molecules, BCS significantly attenuated the release of multiple inflammatory mediators and lipopolysaccharide (LPS)-induced pyroptosis. Its anti-inflammatory mechanism is closely related to the inhibition of intracellular nuclear factor-kappaB (NF-κB) p65 phosphorylation and the noncanonical pyroptosis signalling pathway mediated by caspase-11.
Collapse
Affiliation(s)
- Wenhui Qian
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Bei Zhang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ming Gao
- Department of Pharmacy, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210002, China
| | - Yuting Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jiachen Shen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Dongbing Liang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Chao Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Wei Wei
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Xing Pan
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Qiuying Yan
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing, 210023, China
| | - Dongdong Sun
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing, 210023, China
| | - Dong Zhu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Haibo Cheng
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing, 210023, China
| |
Collapse
|
41
|
Pei X, Ma S, Hong L, Zuo Z, Xu G, Chen C, Shen Y, Liu D, Li C, Li D. Molecular insights of T-2 toxin exposure-induced neurotoxicity and the neuroprotective effect of dimethyl fumarate. Food Chem Toxicol 2025; 196:115166. [PMID: 39617286 DOI: 10.1016/j.fct.2024.115166] [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: 10/27/2024] [Revised: 11/24/2024] [Accepted: 11/28/2024] [Indexed: 12/06/2024]
Abstract
T-2 toxin, a potent environmental pollutant, has been proved to stimulate neuroinflammation, while the connection between T-2 toxin and pyroptosis remain elusive. Dimethyl fumarate (DMF), recently identified as a neuroprotectant and pyroptosis inhibitor, has potential therapeutic applications that are underexplored. Based on present study in vitro and vivo, we demonstrated that T-2 toxin induced the activation of NLRP3-Caspase-1 inflammasome in hippocampal neurons. In addition to proinflammatory mediator overexpression, gasdermin D (GSDMD)-dependently pyroptosis in the mouse hippocampal neuron cell line (HT22) treated by T-2 toxin was determined in our study. Moreover, the palliative effect of knockdown sequence of high mobility group B1 protein (HMGB1) provided more details for T-2 toxin-initiated pyroptosis. Importantly, we confirmed that DMF, as a novel inhibitor of GSDMD, could alleviate pyroptosis induced by T-2 toxin in an GSDMD targeting manner. In summary, our studies exposed the evidence that T-2 toxin could induce NLRP3 inflammasome activation and hippocampal neuronal pyroptosis. More notably, DMF was turn out to be a critical executioner for attenuating GSDMD-mediated pyroptosis. Our data found a new function of DMF and suggested a novel therapy strategy against mycotoxin-triggered neuronal inflammation, which leads to varieties of neurological diseases.
Collapse
Affiliation(s)
- Xingyao Pei
- Open Fund of Key Laboratory of Smart Breeding (Co-construction By Ministry and Province), Ministry of Agriculture and Rural Affairs, Tianjin Agricultural University, Jinjing Road No.22, Xiqing District, Tianjin 300392, China; Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Jinjing Road No.22, Xiqing District, Tianjin 300392, China
| | - Shuhui Ma
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Jinjing Road No.22, Xiqing District, Tianjin 300392, China
| | - Liang Hong
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Jinjing Road No.22, Xiqing District, Tianjin 300392, China
| | - Zonghui Zuo
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Jinjing Road No.22, Xiqing District, Tianjin 300392, China
| | - Gang Xu
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Jinjing Road No.22, Xiqing District, Tianjin 300392, China
| | - Chun Chen
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Jinjing Road No.22, Xiqing District, Tianjin 300392, China
| | - Yao Shen
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Jinjing Road No.22, Xiqing District, Tianjin 300392, China
| | - Dingkuo Liu
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Jinjing Road No.22, Xiqing District, Tianjin 300392, China; Tianjin Key Laboratory of Biological Feed Additive Enterprise, S&E Burgeoning Biotechnology (Tianjin) Co., Ltd, Tianjin 300383, China
| | - Cun Li
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Jinjing Road No.22, Xiqing District, Tianjin 300392, China
| | - Daowen Li
- Open Fund of Key Laboratory of Smart Breeding (Co-construction By Ministry and Province), Ministry of Agriculture and Rural Affairs, Tianjin Agricultural University, Jinjing Road No.22, Xiqing District, Tianjin 300392, China; Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Jinjing Road No.22, Xiqing District, Tianjin 300392, China; Tianjin Key Laboratory of Biological Feed Additive Enterprise, S&E Burgeoning Biotechnology (Tianjin) Co., Ltd, Tianjin 300383, China.
| |
Collapse
|
42
|
Wang K, Shen M, Tang H, Zhou J, Liu Y, Niu D, Zeng Z, Pan L, Yao J, Sun C. Jingfang Granule promotes the tricarboxylic acid cycle to improve chronic fatigue syndrome by increasing the expression of Idh1 and Idh2. JOURNAL OF ETHNOPHARMACOLOGY 2025; 340:119241. [PMID: 39689747 DOI: 10.1016/j.jep.2024.119241] [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: 11/06/2024] [Revised: 12/08/2024] [Accepted: 12/13/2024] [Indexed: 12/19/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Chronic fatigue syndrome (CFS), as a complex, multisystemic, and multisystemic disorder affecting multiple organs and systems, often accompanies by symptoms such as post-exercise discomfort, sleep disorders, cognitive difficulties, and orthostatic intolerance. Jingfang Granule (JFG) is a traditional Chinese medicine that have significant protective effects on CFS, but the mechanism is still vague. AIM OF STUDY This study was designed to evaluate the protective mechanism of JFG on mice with CFS. MATERIALS AND METHODS The combined stimuli method was used to establish the mice CFS model, and JFG was orally administered. The body weight, exhaustion swimming training and tail suspension test were assayed every 7 days to evaluate the improvement of JFG on CFS. Lactic acid, adenosine triphosphate (ATP), malondialdehyde (MDA), superoxide dismutase (SOD), reactive oxygen species (ROS), IL-1β, TNF-α, IL-6 in serum and liver glycogen, muscle glycogen in muscle were analyzed. Transmission electron microscopy was used to detect mitochondrial morphology. The regulatory networks were investigated by proteomics and central carbon metabolomics, which were verified by Western blot. RESULTS JFG reversed the loss of weight and reduce of exhaust swimming time (P < 0.05) induced by CFS in mice, and increased the tail suspension time (P < 0.05), indicating that JFG has an improving effect on CFS. Meanwhile, JFG increased the spleen index (P < 0.05), decreased the thymus index (P < 0.05) and cardiac index (P < 0.05), inhibited the secretion of Lactic acid (P < 0.05), and increased the content of liver glycogen (P < 0.05), muscle glycogen (P < 0.05), and ATP (P < 0.05), and improved mitochondrial morphology in mice with CFS. JFG also inhibited the release of TNF-α (P < 0.05), IL-1β (P < 0.05) and IL-6 (P < 0.05) in serum by inhibiting TLR4/NF-κB signaling pathway and NLRP3 inflammasome signaling pathway, and inhibited oxidative stress by activating Nrf2/HO-1/NQO1 axis. Integrated central carbon metabolomics, proteomics and Western blot showed that JFG intervened in CFS by increasing the expression of Idh1 (P < 0.05) and Idh2 (P < 0.01) to promote tricarboxylic acid (TCA) cycle. CONCLUSIONS This study confirmed that JFG promoted the TCA cycle by increasing the expression of Idh1 and Idh2, and then inhibited inflammation and oxidative stress to prevent CFS injury, which provided a potential drug candidate for CFS treatment.
Collapse
Affiliation(s)
- Kun Wang
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Ji'nan, 250355, China.
| | - Mengmeng Shen
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Hongguang Tang
- College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China.
| | - Jidong Zhou
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Yan Liu
- School of Pharmacy, Qingdao University, Qingdao, 266071, China.
| | - Dejun Niu
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Zhen Zeng
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Lihong Pan
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Jingchun Yao
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Ji'nan, 250355, China; State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China.
| | - Chenghong Sun
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi, 276005, China; College of Food Science and Pharmaceutical Engineering, Zaozhuang University, Zaozhuang, 277160, China.
| |
Collapse
|
43
|
Zhang FL, Chen YL, Luo ZY, Song ZB, Chen Z, Zhang JX, Zheng ZZ, Tan XM. Huashi baidu granule alleviates inflammation and lung edema by suppressing the NLRP3/caspase-1/GSDMD-N pathway and promoting fluid clearance in a porcine reproductive and respiratory syndrome (PRRS) model. JOURNAL OF ETHNOPHARMACOLOGY 2025; 340:119207. [PMID: 39653102 DOI: 10.1016/j.jep.2024.119207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 11/22/2024] [Accepted: 12/02/2024] [Indexed: 12/13/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Huashi Baidu Granule (HSBDG), a traditional Chinese medicine (TCM), is used for treating coronavirus disease 2019 (COVID-19). Porcine reproductive and respiratory syndrome (PRRS) is considered the "COVID-19" for swine. According to the TCM theory, "dampness" is the main pathogenic factor in COVID-19 and PRRS, and "Huashi" means that this formula is good at removing "dampness". Studies have demonstrated that HSBDG's effect in COVID-19; but the mechanism of removing "dampness" remains elusive. AIM OF THE STUDY We aimed to assess the effect of HSBDG on PRRS, and elucidate its potential mechanism in removing "dampness". MATERIALS AND METHODS We established a PRRS-virus (PRRSV)-infected Marc-145 cells model, and performed qRT-PCR, Western blot analysis, and indirect immunofluorescence assay to examine the anti-PRRSV effects of HSBDG in vitro. PRRSV-infected pig model was established and used to investigate HSBDG's effect in PRRS and explore underlying mechanisms in removing "dampness" using ELISA and immunohistochemistry assay methods. RESULTS HSBDG exhibited anti-PRRSV activity and suppressed the viral replication and release phases. HSBDG treatment alleviated PRRS, lowered rectal temperature, reduced histopathological changes and viral load in lung tissues, and ameliorated organ lesions. Moreover, IL-1β, IL-6, IL-8, and TNF-α expressions were decreased in lung tissues. Mechanistically, HSBDG inhibited the NLRP3/Caspase-1/GSDMD-N pathway to reduce the inflammatory response and upregulated AQP1, AQP5, α-ENaC, and Na-K-ATPase expressions to promote lung fluid clearance. CONCLUSION HSBDG exerted anti-PRRSV effects and could attenuate PRRS. HSBDG potentially removes "dampness" by attenuating inflammation by suppressing the NLRP3/Caspase-1/GSDMD-N pathway and inhibiting pulmonary edema by upregulating the expression of AQP1, AQP5, α-ENaC, and Na-K-ATPase.
Collapse
Affiliation(s)
- Feng-Lin Zhang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou, 510515, China.
| | - Yi-Lin Chen
- South China Agricultural University College of Veterinary Medicine, Guangzhou, 510640, China.
| | - Zhen-Ye Luo
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou, 510515, China.
| | - Ze-Bu Song
- South China Agricultural University College of Veterinary Medicine, Guangzhou, 510640, China.
| | - Zhe Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou, 510515, China.
| | - Jia-Xuan Zhang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou, 510515, China.
| | - Ze-Zhong Zheng
- South China Agricultural University College of Veterinary Medicine, Guangzhou, 510640, China.
| | - Xiao-Mei Tan
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, Southern Medical University, Guangzhou, 510515, China; Guangdong Provincial Engineering Laboratory of Chinese Medicine Preparation Technology, Guangzhou, 510515, China.
| |
Collapse
|
44
|
Huang Z, Gu Z, Zeng Y, Zhang D. Platelet-rich plasma alleviates skin photoaging by activating autophagy and inhibiting inflammasome formation. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03800-0. [PMID: 39836253 DOI: 10.1007/s00210-025-03800-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 01/08/2025] [Indexed: 01/22/2025]
Abstract
Platelet-rich plasma (PRP) holds promising prospects for the treatment of skin photoaging. This study aims to unravel the mechanism underlying PRP's anti-photoaging properties. Partial skin of rats was irradiated with ultraviolet (UV) and injected with PRP, and the skin appearance, pathological state, and aging conditions were determined. Apoptosis, reactive oxygen species (ROS), and collagen levels in skin tissues were detected. HaCaT cells were stimulated with UVB, and the effects of PRP on cells and collagen degradation enzymes were evaluated. Furthermore, the mechanism of the autophagy-NLRP3 inflammasome pathway was explored by treating cells with the autophagy inhibitor 3-MA. Erythema, ulceration, and wrinkles appeared on the skin of rats after being irradiated by UV. PRP could enhance skin tenderness and improve skin pathology and aging. PRP inhibited cell apoptosis, ROS generation, and collagen degradation in skin tissue. PRP elevated UVB-stimulated HaCaT cell activity, reduced oxidative stress, senescence, and MMP-1. Furthermore, 3-MA treatment reversed the inhibition of NLRP3 inflammasome by PRP, suggesting that autophagy mediated the regulation of PRP. To summarize, this study elucidates the regulatory mechanism of PRP on the autophagy-NLRP3 inflammasome pathway in the photoaging. These findings may provide a novel theoretical foundation for the clinical application of PRP.
Collapse
Affiliation(s)
- Zuocai Huang
- Department of Emergency Medicine, Dongshan Hospital, Meijiang District, Meizhou, 514011, Guangdong, China
| | - Zhonghang Gu
- Department of Dermatology, Dongshan Hospital, Guofengyuan Building, Xuezi Avenue, Meijiang District, Meizhou, 514011, Guangdong, China
| | - Yuanjuan Zeng
- Department of Dermatology, Meizhou People's Hospital, Meijiang District, Meizhou, 514011, Guangdong, China
| | - Dongxing Zhang
- Department of Dermatology, Dongshan Hospital, Guofengyuan Building, Xuezi Avenue, Meijiang District, Meizhou, 514011, Guangdong, China.
| |
Collapse
|
45
|
Cambon A, Guervilly C, Delteil C, Potere N, Bachelier R, Tellier E, Abdili E, Leprince M, Giani M, Polidoro I, Albanese V, Ferrante P, Coffin L, Schiffrin M, Arnaud L, Lacroix R, Roque S, Forel JM, Hraiech S, Daniel L, Papazian L, Dignat-George F, Kaplanski G. Caspase-1 activation, IL-1/IL-6 signature and IFNγ-induced chemokines in lungs of COVID-19 patients. Front Immunol 2025; 15:1493306. [PMID: 39882243 PMCID: PMC11774885 DOI: 10.3389/fimmu.2024.1493306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 12/11/2024] [Indexed: 01/31/2025] Open
Abstract
Rationale COVID-19-associated acute-respiratory distress syndrome (C-ARDS) results from a direct viral injury associated with host excessive innate immune response mainly affecting the lungs. However, cytokine profile in the lung compartment of C-ARDS patients has not been widely studied, nor compared to non-COVID related ARDS (NC-ARDS). Objectives To evaluate caspase-1 activation, IL-1 signature, and other inflammatory cytokine pathways associated with tissue damage using post-mortem lung tissues, bronchoalveolar lavage fluids (BALF), and serum across the spectrum of COVID-19 severity. Methods Histological features were described and activated-caspase-1 labeling was performed in 40 post-mortem biopsies. Inflammatory cytokines were quantified in BALF and serum from 19 steroid-treated-C-ARDSand compared to 19 NC-ARDS. Cytokine concentrations were also measured in serum from 128 COVID-19 patients at different severity stages. Measurements and main results Typical "diffuse alveolar damage" in lung biopsies were associated with activated caspase-1 expression and vascular lesions. Soluble Caspase-1p20, IL-1β, IL-1Ra, IL-6 and at lower level IFNγ and CXCL-10, were highly elevated in BALF from steroid-treated-C-ARDS as well as in NC-ARDS. IL-1β appeared concentrated in BALF, whereas circulating IL-6 and IL-1Ra concentrations were comparable to those in BALF and correlated with severity. TNFα, TNFR1 and CXCL8 however, were significantly higher in NC-ARDS compared to C-ARDS, treated by steroid. Conclusions In the lungs of C-ARDS, both caspase-1 activation with a predominant IL-1β/IL-6 signature and IFNγ -associated chemokines are elevated despite steroid treatment. These pathways may be specifically targeted in ARDS to improve response to treatment and to limit alveolar and vascular lung damage.
Collapse
Affiliation(s)
- Audrey Cambon
- Aix-Marseille Université, INSERM, INRAE, C2VN, Marseille, France
| | - Christophe Guervilly
- Centre d’Etudes et de Recherches sur les Services de Santé et qualité de vie EA 3279, Aix-Marseille Université, Marseille, France
- Service de Médecine Intensive Réanimation, Hôpital Nord, Assistance Publique- Hôpitaux de Marseille, Chemin des Bourrely, Marseille, France
| | - Clémence Delteil
- Département de Médecine légale, Hôpital de la Timone, Assistance Publique-Hôpitaux de Marseille, Marseille University, Marseille, France
| | - Nicola Potere
- School of Medicine and Health Sciences, “G. d’Annunzio” University of Chieti-Pescara, Chieti, Italy
| | | | - Edwige Tellier
- Aix-Marseille Université, INSERM, INRAE, C2VN, Marseille, France
| | - Evelyne Abdili
- Aix-Marseille Université, INSERM, INRAE, C2VN, Marseille, France
- Service d’Hématologie et de Biologie vasculaire, CHU La Timone, APHM, Marseille, France
| | - Marine Leprince
- Service de Médecine interne et d’Immunologie clinique, Assistance Publique - Hôpitaux de Marseille, Hôpital La Conception, Marseille, France
| | - Marco Giani
- School of Medicine and Health Sciences, “G. d’Annunzio” University of Chieti-Pescara, Chieti, Italy
| | - Ildo Polidoro
- Unit of Legal Medicine, “Santo Spirito” Hospital, Local Health Authority of Pescara, Pescara, Italy
| | - Valentina Albanese
- Unit of Legal Medicine, “Santo Spirito” Hospital, Local Health Authority of Pescara, Pescara, Italy
| | - Paolo Ferrante
- Unit of Legal Medicine, “Santo Spirito” Hospital, Local Health Authority of Pescara, Pescara, Italy
| | | | | | - Laurent Arnaud
- Service d’Hématologie et de Biologie vasculaire, CHU La Timone, APHM, Marseille, France
| | - Romaric Lacroix
- Aix-Marseille Université, INSERM, INRAE, C2VN, Marseille, France
- Service d’Hématologie et de Biologie vasculaire, CHU La Timone, APHM, Marseille, France
| | - Sandrine Roque
- Service de Médecine interne et d’Immunologie clinique, Assistance Publique - Hôpitaux de Marseille, Hôpital La Conception, Marseille, France
| | - Jean-Marie Forel
- Centre d’Etudes et de Recherches sur les Services de Santé et qualité de vie EA 3279, Aix-Marseille Université, Marseille, France
- Service de Médecine Intensive Réanimation, Hôpital Nord, Assistance Publique- Hôpitaux de Marseille, Chemin des Bourrely, Marseille, France
| | - Sami Hraiech
- Centre d’Etudes et de Recherches sur les Services de Santé et qualité de vie EA 3279, Aix-Marseille Université, Marseille, France
- Service de Médecine Intensive Réanimation, Hôpital Nord, Assistance Publique- Hôpitaux de Marseille, Chemin des Bourrely, Marseille, France
| | - Laurent Daniel
- Service d’Anatomopathologie, APHM, Aix Marseille University, Marseille, France
| | - Laurent Papazian
- Service de Réanimation, Centre Hospitalier de Bastia, Bastia, France
| | - Françoise Dignat-George
- Aix-Marseille Université, INSERM, INRAE, C2VN, Marseille, France
- Service d’Hématologie et de Biologie vasculaire, CHU La Timone, APHM, Marseille, France
| | - Gilles Kaplanski
- Aix-Marseille Université, INSERM, INRAE, C2VN, Marseille, France
- Service de Médecine interne et d’Immunologie clinique, Assistance Publique - Hôpitaux de Marseille, Hôpital La Conception, Marseille, France
| |
Collapse
|
46
|
Li D, Dai Y, Li Z, Bi H, Li H, Wang Y, Liu Y, Tian X, Chen L. Resveratrol Upregulates miR-124-3p Expression to Target DAPK1, Regulating the NLRP3/Caspase-1/GSDMD Pathway to Inhibit Pyroptosis and Alleviate Spinal Cord Injury. J Cell Mol Med 2025; 29:e70338. [PMID: 39833100 PMCID: PMC11745821 DOI: 10.1111/jcmm.70338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 11/29/2024] [Accepted: 12/23/2024] [Indexed: 01/30/2025] Open
Abstract
Currently, an effective treatment for spinal cord injury (SCI) is not available. Due to the irreversible primary injury associated with SCI, the prevention and treatment of secondary injury are very important. In the secondary injury stage, pyroptosis exacerbates the deterioration of the spinal cord injury, and inhibiting pyroptosis is beneficial for recovery from SCI. The aim of this study was to clarify the role of resveratrol (RES) and the antipyroptotic mechanisms of RES and miR-124-3p in SCI to lay a theoretical foundation for the clinical treatment of SCI and provide new therapeutic approaches. Using cell staining and related molecular protein detection techniques to assess DAPK1, the effects of miR-124-3p and RES on pyroptosis were investigated, and the effects of RES on injured spinal cord repair in rats were evaluated using tissue staining and related functional recovery experiments. In vitro, DAPK1 interacts with NLRP3, exerting a pyroptotic effect through the NLRP3/Caspase-1/GSDMD pathway and DAPK1 knockdown inhibits pyroptosis. miR-124-3P negatively regulates the level of DAPK1 and reduced cell pyroptosis. RES increased miR-124-3p expression and reduces DAPK1 expression, affecting the NLRP3/Caspase-1/GSDMD pathway and inhibiting pyroptosis. In vivo, RES reduces GSDMD-N levels in rats with SCI, promotes functional recovery, and thus promotes recovery from SCI. Therefore, we concluded that RES increases the level of miR-124-3p, which targets DAPK1, regulates the NLRP3/Caspase-1/GSDMD pathway, inhibits pyroptosis and alleviates SCI.
Collapse
Affiliation(s)
- Daohui Li
- Department of OrthopedicsThe First Affiliated Hospital of Kunming Medical UniversityYunnanChina
| | - Yongwen Dai
- Department of OrthopedicsThe First Affiliated Hospital of Kunming Medical UniversityYunnanChina
| | - Zhengtao Li
- Department of OrthopedicsThe First Affiliated Hospital of Kunming Medical UniversityYunnanChina
| | - Hangchuan Bi
- Department of OrthopedicsThe First Affiliated Hospital of Kunming Medical UniversityYunnanChina
| | - Haotian Li
- Department of OrthopedicsThe First Affiliated Hospital of Kunming Medical UniversityYunnanChina
| | - Yongquan Wang
- Department of OrthopedicsThe First Affiliated Hospital of Kunming Medical UniversityYunnanChina
| | - Yuan Liu
- Department of OrthopedicsThe First Affiliated Hospital of Kunming Medical UniversityYunnanChina
| | - Xinpeng Tian
- Department of Critical Care MedicineXi Chang People's HospitalSichuanChina
| | - Lingqiang Chen
- Department of OrthopedicsThe First Affiliated Hospital of Kunming Medical UniversityYunnanChina
| |
Collapse
|
47
|
Zheng Y, Wu Z, Wei X, Zhang L, Hu Y, Zhou Z. 1,25(OH)2D3 promotes insulin secretion through the classical pyroptosis pathway in vitro and vivo. Biochem Biophys Res Commun 2025; 742:151058. [PMID: 39642712 DOI: 10.1016/j.bbrc.2024.151058] [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: 11/14/2024] [Accepted: 11/22/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND Diabetes is a chronic metabolic disorder characterized by persistently elevated levels of blood glucose. Research has demonstrated a close relationship between inflammation and the development of diabetes. Vitamin D has been shown to be significantly associated with type 2 diabetes; however, the mechanisms by which it regulates inflammation during the onset of the disease remain incompletely understood. In this study, we investigated the effect of pyroptosis on pancreatic β-cell function in diabetes and explored the role of 1,25(OH)2D3 in type 2 diabetes through the pyroptosis signaling pathway. METHODS In both in vivo and in vitro settings, we established a diabetes model combined with 1,25(OH)₂D₃ intervention to investigate its impact on insulin secretion levels, the release of inflammatory factors, and the expression levels of pyroptosis-related proteins. RESULTS In both in vivo and in vitro experiments, we have observed that 1,25(OH)₂D₃ exhibits anti-inflammatory properties by downregulating the expression levels of pyroptosis-related proteins. Furthermore, it provides protection against pancreatic β-cell damage caused by type 2 diabetes mellitus (T2DM) and enhances insulin secretion. Inhibition of gasdermin D (GSDMD) expression impedes the progression of cell pyroptosis, reduces the amplification of the inflammatory response, and protects pancreatic cells from injury. CONCLUSION We hypothesize that the induction of pancreatic cells through pyroptosis occurs via the classical pathway in T2DM, and propose that 1,25(OH)2D3 may have a beneficial effect on this process. Consequently, 1,25(OH)2D3 could potentially serve as an adjuvant to inhibit the pyroptosis of pancreatic β cells by targeting the classical signaling pathway, thereby reducing the inflammatory response and alleviating symptoms associated with diabetes.
Collapse
Affiliation(s)
- Yuxuan Zheng
- Laboratory Animal Center of Suzhou Medical College, Soochow University, Suzhou, China
| | - Zhihao Wu
- Laboratory Animal Center of Suzhou Medical College, Soochow University, Suzhou, China
| | - Xun Wei
- Center of Laboratory Animal, Shanghai Jiao Tong University, Shanghai, China
| | - Lewen Zhang
- Laboratory Animal Center of Suzhou Medical College, Soochow University, Suzhou, China
| | - Yudie Hu
- Laboratory Animal Center of Suzhou Medical College, Soochow University, Suzhou, China
| | - Zhengyu Zhou
- Laboratory Animal Center of Suzhou Medical College, Soochow University, Suzhou, China.
| |
Collapse
|
48
|
Ling L, Li R, Xu M, Zhou J, Hu M, Zhang X, Zhang XJ. Species differences of fatty liver diseases: comparisons between human and feline. Am J Physiol Endocrinol Metab 2025; 328:E46-E61. [PMID: 39636211 DOI: 10.1152/ajpendo.00014.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 11/25/2024] [Accepted: 11/25/2024] [Indexed: 12/07/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) has emerged as the most widespread chronic liver disease that poses significant threats to public health due to changes in dietary habits and lifestyle patterns. The transition from simple steatosis to nonalcoholic steatohepatitis (NASH) markedly increases the risk of developing cirrhosis, hepatocellular carcinoma, and liver failure in patients. However, there is only one Food and Drug Administration-approved therapeutic drug in the world, and the clinical demand is huge. There is significant clinical heterogeneity among patients with NAFLD, and it is challenging to fully understand human NAFLD using only a single animal model. Interestingly, felines, like humans, are particularly prone to spontaneous fatty liver disease. This review summarized and compared the etiology, clinical features, pathological characteristics, and molecular pathogenesis between human fatty liver and feline hepatic lipidosis (FHL). We analyzed the key similarities and differences between those two species, aiming to provide theoretical foundations for developing effective strategies for the treatment of NAFLD in clinics.
Collapse
Affiliation(s)
- Like Ling
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China
| | - Ruilin Li
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China
| | - Mengqiong Xu
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China
| | - Junjie Zhou
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China
| | - Manli Hu
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China
| | - Xin Zhang
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China
| | - Xiao-Jing Zhang
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Gannan Innovation and Translational Medicine Research Institute, Gannan Medical University, Ganzhou, China
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
| |
Collapse
|
49
|
Yao Q, Wei T, Qiu H, Cai Y, Yuan L, Liu X, Li X. Epigenetic Effects of Natural Products in Inflammatory Diseases: Recent Findings. Phytother Res 2025; 39:90-137. [PMID: 39513382 DOI: 10.1002/ptr.8364] [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: 03/06/2024] [Revised: 06/17/2024] [Accepted: 07/12/2024] [Indexed: 11/15/2024]
Abstract
Inflammation is an essential step for the etiology of multiple diseases. Clinically, due to the limitations of current drugs for the treatment of inflammatory diseases, such as serious side effects and expensive costs, it is urgent to explore novel mechanisms and medicines. Natural products have received extensive attention recently because of their multi-component and multi-target characteristics. Epigenetic modifications are crucial pathophysiological targets for developing innovative therapies for pharmacological interventions. Investigations examining how natural products improving inflammation through epigenetic modifications are emerging. This review state that natural products relieve inflammation via regulating the gene transcription levels through chromosome structure regulated by histone acetylation levels and the addition or deletion of methyl groups on DNA duplex. They could also exert anti-inflammatory effects by modulating the proteins in typical inflammatory signaling pathways by ubiquitin-related degradation and the effect of glycolysis derived free glycosyls. Studies on epigenetic modifications have the potential to facilitate the development of natural products as therapeutic agents. Future research directed at better understanding of how natural products modulate inflammatory processes through less studied epigenetic modifications including neddylation, SUMOylation, palmitoylation and lactylation, may provide new implications. Meanwhile, higher quality preclinical studies and more powerful clinical evidence are still needed to firmly establish the clinical efficacy of the natural products. Trial Registration: ClinicalTrials.gov Identifier: NCT01764204; ClinicalTrials.gov Identifier: NCT05845931; ClinicalTrials.gov Identifier: NCT04657926; ClinicalTrials.gov Identifier: NCT02330276.
Collapse
Affiliation(s)
- Qianyi Yao
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing, China
| | - Tanjun Wei
- Department of Pharmacy, Dazhou Integrated TCM & Western Medical Hospital, Sichuan, China
| | - Hongmei Qiu
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing, China
| | - Yongqing Cai
- Department of Pharmacy, Daping Hospital, Army Medical University, Chongqing, China
| | - Lie Yuan
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing, China
| | - Xin Liu
- Medical Research Center, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Xiaoli Li
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, China
- Key Laboratory for Biochemistry and Molecular Pharmacology of Chongqing, Chongqing, China
| |
Collapse
|
50
|
Fan X, Sun L, Qin Y, Liu Y, Wu S, Du L. The Role of HSP90 Molecular Chaperones in Depression: Potential Mechanisms. Mol Neurobiol 2025; 62:708-717. [PMID: 38896156 DOI: 10.1007/s12035-024-04284-4] [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: 04/28/2024] [Accepted: 06/05/2024] [Indexed: 06/21/2024]
Abstract
Major depressive disorder (MDD) is characterized by high rates of disability and death and has become a public health problem that threatens human life and health worldwide. HPA axis disorder and neuroinflammation are two common biological abnormalities in MDD patients. Hsp90 is an important molecular chaperone that is widely distributed in the organism. Hsp90 binds to the co-chaperone and goes through a molecular chaperone cycle to complete its regulation of the client protein. Numerous studies have demonstrated that Hsp90 regulates how the HPA axis reacts to stress and how GR, the HPA axis' responsive substrate, matures. In addition, Hsp90 exhibits pro-inflammatory effects that are closely related to neuroinflammation in MDD. Currently, Hsp90 inhibitors have made some progress in the treatment of a variety of human diseases, but they still need to be improved. Further insight into the role of Hsp90 in MDD provides new ideas for the development of new antidepressant drugs targeting Hsp90.
Collapse
Affiliation(s)
- Xuyuan Fan
- Department of Medicine, Yangzhou University, Yangzhou, 225012, Jiangsu, China
| | - Lei Sun
- Department of Medicine, Yangzhou University, Yangzhou, 225012, Jiangsu, China
| | - Ye Qin
- Department of Laboratory Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225012, Jiangsu, China
| | - Yuan Liu
- Department of Laboratory Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225012, Jiangsu, China
| | - Shusheng Wu
- Department of the Central Laboratory, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225012, Jiangsu, China.
| | - Longfei Du
- Department of Laboratory Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225012, Jiangsu, China.
| |
Collapse
|