1
|
Lin H, Lv Y, Liu H, Lin Z, Zhu N, Huang B. Baicalein alleviates chronic acute stress-induced irritable bowel syndrome-like symptoms in rats via modulating the ODC1/NF-κB pathway and oxidative stress. Biochem Biophys Res Commun 2025; 771:152058. [PMID: 40409117 DOI: 10.1016/j.bbrc.2025.152058] [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: 02/13/2025] [Revised: 04/14/2025] [Accepted: 05/19/2025] [Indexed: 05/25/2025]
Abstract
BACKGROUND As a persistent gastrointestinal disorder, irritable bowel syndrome (IBS) influences the gut-brain connection, leading to abdominal pain and altered bowel irregularities. Baicalein, a flavonoid extracted from Scutellaria baicalensis, is frequently utilized in anti-inflammatory treatments. This study aimed to explore baicalein's effectiveness in mitigating IBS symptoms triggered by both chronic and acute stress (CAS) and to uncover its fundamental mechanisms. METHODS Sprague-Dawley rats were employed to develop an IBS rat model by inducing CAS for five weeks. Each rat was randomly allocated to one of four experimental groups: model (M), low-dose baicalein (L), high-dose baicalein (H), and control (C). Baicalein was orally administered throughout the experiment. Behavioral assessments were conducted, including forced swimming, marble-burying, intestinal motility, and visceral sensitivity tests. Colonic tissues were collected for histopathological examination, evaluation of oxidative stress (MDA and SOD levels), and analysis of inflammatory cytokines, and ODC1/NF-κB pathway activation using Western blot assay, ELISA, and immunofluorescence. RESULTS Baicalein treatment notably ameliorated IBS-like symptoms, such as fecal pellet output and AWR scores, by alleviating stress-induced behavioral changes. Baicalein bolstered antioxidant defenses through boosting SOD activity and lowering MDA levels. Moreover, baicalein inhibited inflammatory responses, targeting IL-6, IL-1β, and TNF-α, while suppressing the expression of ODC1 and restraining NF-κB p65 phosphorylation within the colon. These results indicate that baicalein modulates oxidative stress and inflammation in CAS-associated IBS. CONCLUSION Baicalein demonstrates protection against CAS-induced IBS by diminishing intestinal inflammation, oxidative damage, and visceral hypersensitivity via suppression of the ODC1/NF-κB pathway. These findings underscore baicalein's potential as a therapeutic approach for IBS.
Collapse
Affiliation(s)
- Han Lin
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, People's Republic of China
| | - Yunlong Lv
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, People's Republic of China
| | - Huacheng Liu
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, People's Republic of China
| | - Zheng Lin
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, People's Republic of China
| | - Ning Zhu
- Department of Cardiology, The Third Affiliated Hospital of Shanghai University (Wenzhou People's Hospital), The Wenzhou Third Clinical Institute Affiliated to Wenzhou Medical University, No. 299 Guan Road, Wenzhou, 325000, Zhejiang Province, People's Republic of China.
| | - Bingwu Huang
- Department of Anesthesiology and Perioperative Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, People's Republic of China.
| |
Collapse
|
2
|
Huang Y, Qiu H, Chen W, Meng Z, Cai Y, Qiao D, Yue X. Identification of TRAF2, CAMK2G, and TIMM17A as biomarkers distinguishing mechanical asphyxia from sudden cardiac death base on 4D-DIA Proteomics: A pilot study. J Pharm Biomed Anal 2025; 258:116730. [PMID: 39921950 DOI: 10.1016/j.jpba.2025.116730] [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: 11/03/2024] [Revised: 01/10/2025] [Accepted: 02/02/2025] [Indexed: 02/10/2025]
Abstract
In the context of forensic medicine, the differential diagnosis between mechanical asphyxia and sudden cardiac death is very important regarding the establishment of the cause of death. Traditional autopsy findings have generally been very nonspecific; accordingly, highlighting the need for more specific molecular biomarkers. This study employed four-dimensional data-independent acquisition (4D-DIA) proteomics technology, in combination with both animal models and human samples, to conduct a comprehensive protein expression analysis of cardiac tissues, identifying 7557 proteins, among which 142 shared differentially expressed proteins (DEPS) were screened out. Based on the protein interaction network and through rigorous screening, this study identified three proteins, namely TNF receptor-associated factor 2 (TRAF2), Calcium/calmodulin-dependent protein kinase II gamma (CAMK2G), and translocase of inner mitochondrial membrane 17 homolog A (TIMM17A), as biomarkers for differentiating mechanical asphyxia from sudden cardiac death. Further verification using Western Blot (WB) and immunohistochemistry (IHC) proved the differential expression of these biomarkers in both animal and human samples. These findings, besides deepening the molecular understanding of the pathophysiological differences between sudden cardiac death and mechanical asphyxia, also provided new biomarkers for forensic applications that could enable the improvement of accuracy and reliability in the determination of the cause of death.
Collapse
Affiliation(s)
- Yuebing Huang
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Hai Qiu
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Wen Chen
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Zilin Meng
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yu Cai
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Dongfang Qiao
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Xia Yue
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China.
| |
Collapse
|
3
|
Askarizadeh F, Butler AE, Kesharwani P, Sahebkar A. Regulatory effect of curcumin on CD40:CD40L interaction and therapeutic implications. Food Chem Toxicol 2025; 200:115369. [PMID: 40043936 DOI: 10.1016/j.fct.2025.115369] [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] [Revised: 01/24/2025] [Accepted: 03/02/2025] [Indexed: 04/21/2025]
Abstract
Natural compounds have garnered significant attention as potential therapeutic agents due to their inherent properties. Their notable qualities, including safety, efficacy, favorable pharmacokinetic properties, and heightened effectiveness against certain diseases, particularly inflammatory conditions, make them particularly appealing. Among these compounds, curcumin has attracted considerable interest for its unique therapeutic properties and has therefore been extensively studied as a potential therapeutic agent for treating various diseases. Curcumin exhibits diverse anti-inflammatory, antioxidant, and antimicrobial effects. Curcumin's immune system regulatory ability has made it a promising compound for treatment of various inflammatory diseases, such as psoriasis, atherosclerosis, asthma, colitis, IBD, and arthritis. Among the signaling pathways implicated in these conditions, the CD40 receptor together with its ligand, CD40L, are recognized as central players. Studies have demonstrated that the interaction between CD40 and CD40L interaction acts as the primary mediator of the immune response in inflammatory diseases. Numerous studies have explored the impact of curcumin on the CD40:CD40L pathway, highlighting its regulatory effects on this inflammatory pathway and its potential therapeutic use in related inflammatory conditions. In this review, we will consider the evidence concerning curcumin's modulatory effects in inflammatory disease and its potential therapeutic role in regulating the CD40:CD40L pathway.
Collapse
Affiliation(s)
- Fatemeh Askarizadeh
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India.
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Institute of Medical and Technical Sciences, Saveetha Medical College and Hospitals, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
4
|
Wu F, Deng Y, Sokolov EP, Falfushynska H, Glänzer A, Xie L, Sokolova IM. Nanopollutants (nZnO) amplify hypoxia-induced cellular stress in a keystone marine bivalve, Mytilus edulis. ENVIRONMENTAL RESEARCH 2025; 274:121346. [PMID: 40058547 DOI: 10.1016/j.envres.2025.121346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 02/18/2025] [Accepted: 03/06/2025] [Indexed: 03/14/2025]
Abstract
Zinc oxide nanoparticles (nZnO) are increasingly utilized in industrial, medical, and personal care products, particularly as the main ingredient in sunscreens, raising concerns about their environmental impact, especially in coastal ecosystems. The Baltic Sea, experiencing severe eutrophication, faces persistent hypoxia due to excessive nutrient runoff and limited water exchange. Simultaneously, coastal pollution from industrial and urban activities introduces nZnO, a highly biotoxic nanopollutant. The combined effects of hypoxia and nZnO contamination may amplify environmental stress, yet their interactions remain insufficiently studied. This study investigates the combined effects of nZnO exposure and fluctuating dissolved oxygen regimes (specifically short- and long-term hypoxia and subsequent reoxygenation) on Mytilus edulis, a sentinel species in these ecosystems. By assessing a range of cellular and molecular markers, including oxidative stress, oxygen sensing, protein quality control, stress response, apoptosis, and inflammation, we show that nZnO exacerbates hypoxia-induced oxidative stress, delaying redox recovery and prolonging oxidative damage during reoxygenation. Specifically, nZnO exposure maintains elevated LPO and PC levels after reoxygenation, indicating prolonged oxidative imbalance. While M. edulis typically recovers from hypoxia-induced stress, nZnO disrupts this process by impairing antioxidant defenses, prolonging HIF-1α activation, and dysregulating p53, JNK, and p38 expression, thereby interfering with normal hypoxia-reoxygenation response. Additionally, nZnO alters HSP70, Lon protease, and caspase-3 regulation, disrupting protein-folding and apoptotic pathways. These findings suggest a synergistic interaction between nZnO and hypoxia, heightening the organism's vulnerability to environmental stress and suggesting risks for marine organisms in nanoparticle-polluted, hypoxia-prone coastal regions.
Collapse
Affiliation(s)
- Fangli Wu
- School of Environment, South China Normal University, University Town, Guangzhou, 510006, China; Department of Marine Biology, Institute for Biological Sciences, University of Rostock, Rostock, Germany; SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou, 510006, China; Fujian Key Laboratory on Conservation and Sustainable Utilization of Marine Biodiversity, Minjiang University, Fuzhou, China
| | - Yuqing Deng
- School of Environment, South China Normal University, University Town, Guangzhou, 510006, China; SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou, 510006, China
| | - Eugene P Sokolov
- Department of Marine Biology, Institute for Biological Sciences, University of Rostock, Rostock, Germany
| | - Halina Falfushynska
- Department of Marine Biology, Institute for Biological Sciences, University of Rostock, Rostock, Germany; Faculty of Economics, Anhalt University of Applied Sciences, 06406, Köthen, Germany; ENERTRAG SE, Gut Dauerthal, Dauerthal, 17291, Germany
| | - Aneka Glänzer
- Department of Marine Biology, Institute for Biological Sciences, University of Rostock, Rostock, Germany
| | - Lingtian Xie
- School of Environment, South China Normal University, University Town, Guangzhou, 510006, China; SCNU Environmental Research Institute, Guangdong Provincial Key Laboratory of Chemical Pollution and Environmental Safety & MOE Key Laboratory of Theoretical Chemistry of Environment, South China Normal University, Guangzhou, 510006, China
| | - Inna M Sokolova
- Department of Marine Biology, Institute for Biological Sciences, University of Rostock, Rostock, Germany; Department of Maritime Systems, Interdisciplinary Faculty, University of Rostock, Rostock, Germany.
| |
Collapse
|
5
|
Li A, Luo M, Liu X, Wu H, Liu X, Zhang Z, Zhang X. Toll-like receptor 3 activation enhances antitumor immune response in lung adenocarcinoma through NF-κB signaling pathway. Front Immunol 2025; 16:1585747. [PMID: 40406122 PMCID: PMC12095255 DOI: 10.3389/fimmu.2025.1585747] [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: 03/01/2025] [Accepted: 04/21/2025] [Indexed: 05/26/2025] Open
Abstract
Background Toll-like receptor 3 (TLR3) is a pattern recognition receptor known to play a crucial role in the immune response to cancer. However, its effect on the efficacy of immunotherapy in lung adenocarcinoma (LUAD) remains unclear. This study aims to investigate the role of TLR3 in LUAD by examining its expression levels, prognostic significance, and impact on immune signaling pathways. Methods We analyzed the impact of TLR3 expression on the prognosis of lung adenocarcinoma patients using data from the Cancer Genome Atlas (TCGA) database and four additional cohorts (GSE72094, GSE30219, GSE50081 and GSE31210). Functional enrichment analyses were performed to compare molecular features between low and high TLR3 expression groups using gene set variation analysis (GSVA). We also examined the correlation between TLR3 and tumor mutation burden (TMB), immune infiltration, and PD-L1 expression. Further experimental validation was conducted using co-culture systems of LUAD cells and peripheral blood mononuclear cells (PBMCs) with PD1 inhibitors, and Western blot analysis to investigate the involvement of NF-κB signaling. Results TLR3 expression was significantly lower in LUAD tissues compared to normal tissues, with high TLR3 expression correlating with better survival outcomes across multiple cohorts. High TLR3 expression was associated with increased TMB and enhanced immune activation. Patients with high TLR3 expression exhibited higher immune checkpoint expression and immune cell infiltration. Experimental results showed that TLR3 agonists increased the susceptibility of LUAD cells to activated PBMCs under PD1 inhibitor therapy, inhibiting cell proliferation, migration, and invasion. Additionally, TLR3 has a strong positive correlation with MHC molecules and upregulated PD-L1 expression. NF-κB was identified as a key regulator of PD-L1 expression, with TLR3 agonists enhancing NF-κB and PD-L1 activity. Conclusion TLR3 enhances the anti-tumor immune response in LUAD by modulating NF-κB signaling and PD-L1 expression, making it a promising prognostic biomarker and therapeutic target. This study highlights the potential of TLR3 to improve immunotherapy outcomes, providing a comprehensive analysis of its role in LUAD and paving the way for novel therapeutic strategies targeting TLR3-mediated pathways.
Collapse
Affiliation(s)
- Ang Li
- School of Public Health, North China University of Science and Technology, Tangshan, China
- College of Life Science, North China University of Science and Technology, Tangshan, China
- Hebei Key Laboratory of Occupational Health and Safety for Coal Industry, North China University of Science and Technology, Tangshan, China
| | - Man Luo
- College of Life Science, North China University of Science and Technology, Tangshan, China
| | - Xiyao Liu
- College of Life Science, North China University of Science and Technology, Tangshan, China
| | - Hongjiao Wu
- School of Public Health, North China University of Science and Technology, Tangshan, China
- College of Life Science, North China University of Science and Technology, Tangshan, China
- Hebei Key Laboratory of Occupational Health and Safety for Coal Industry, North China University of Science and Technology, Tangshan, China
| | - Xiaoguang Liu
- College of Life Science, North China University of Science and Technology, Tangshan, China
| | - Zhi Zhang
- Affliated Tangshan Gongren Hospital, North China University of Science and Technology, Tangshan, China
| | - Xuemei Zhang
- School of Public Health, North China University of Science and Technology, Tangshan, China
- College of Life Science, North China University of Science and Technology, Tangshan, China
- Hebei Key Laboratory of Occupational Health and Safety for Coal Industry, North China University of Science and Technology, Tangshan, China
| |
Collapse
|
6
|
Jin J, Dong Y, Huang Y, Wu L, Yu L, Sun Y, Zhou Q, Yin HY, Gu WJ. ERRα Knockout Promotes M2 Microglial Polarization and Inhibits Ferroptosis in Sepsis-Associated Brain Dysfunction. Mol Neurobiol 2025:10.1007/s12035-025-05005-1. [PMID: 40325331 DOI: 10.1007/s12035-025-05005-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Accepted: 04/29/2025] [Indexed: 05/07/2025]
Abstract
Sepsis-associated brain dysfunction (SABD) is a critical neurological complication with high mortality, yet its pathogenesis remains poorly understood. This study investigated the role of estrogen-related receptor α (ERRα) in SABD pathogenesis using ERRα knockout (KO) mice and cecal ligation and puncture (CLP) models. We found that ERRα KO mice exhibited improved survival rates, milder neurological symptoms, reduced pro-inflammatory cytokine production (TNF-α, IL-1β), and increased anti-inflammatory cytokine (IL-10) levels compared to wild-type controls. Additionally, ERRα deficiency promoted microglial M2 polarization and attenuated ferroptosis, as evidenced by decreased iron accumulation, reduced lipid peroxidation, and normalized mitochondrial morphology. Mechanistically, these protective effects were mediated through inhibition of the NF-κB signaling pathway. In vitro studies with ERRα knockdown in LPS-stimulated BV2 microglia confirmed these findings. Our results suggest that ERRα as a critical regulator of microglial function in SABD through coordinated control of inflammatory responses, polarization states, and ferroptosis, suggesting that targeting ERRα may represent a promising therapeutic strategy for SABD treatment.
Collapse
Affiliation(s)
- Jun Jin
- Department of Intensive Care Unit, The First Affiliated Hospital of Jinan University, 613 Huangpu Avenue West, Guangzhou, 510630, China
- Department of Intensive Care Unit, The University of Hong Kong-Shenzhen Hospital, Haiyuan 1St Road, Futian District, Shenzhen, 518000, China
| | - Yang Dong
- Department of Obstetrics and Gynecology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Shenzhen Huarui Model Organisms Biotechnology Co., LTD, Shenzhen, China
| | - Yu Huang
- Department of Intensive Care Unit, The First Affiliated Hospital of Jinan University, 613 Huangpu Avenue West, Guangzhou, 510630, China
| | - Lili Wu
- Department of Intensive Care Unit, The University of Hong Kong-Shenzhen Hospital, Haiyuan 1St Road, Futian District, Shenzhen, 518000, China
| | - Lei Yu
- Department of Intensive Care Unit, The University of Hong Kong-Shenzhen Hospital, Haiyuan 1St Road, Futian District, Shenzhen, 518000, China
| | - Yuanyuan Sun
- Department of Medical Genetics, China Medical University, Shenyang, China
| | - Qingshan Zhou
- Department of Intensive Care Unit, The University of Hong Kong-Shenzhen Hospital, Haiyuan 1St Road, Futian District, Shenzhen, 518000, China.
| | - Hai-Yan Yin
- Department of Intensive Care Unit, The First Affiliated Hospital of Jinan University, 613 Huangpu Avenue West, Guangzhou, 510630, China.
| | - Wan-Jie Gu
- Department of Intensive Care Unit, The First Affiliated Hospital of Jinan University, 613 Huangpu Avenue West, Guangzhou, 510630, China.
| |
Collapse
|
7
|
Chen JQ, Wu XJ, Wu XX, Geng BD, Zhou D, Wen J, Chan SCL, Jin C, Xu JW, Lu JH, Ge G. Protective effect of aqueous extract of Reineckea carnea (Andrews) Kunth against cigarette smoke-induced chronic obstructive pulmonary disease in mice and its impact on gut microbiota. Fitoterapia 2025; 184:106600. [PMID: 40339613 DOI: 10.1016/j.fitote.2025.106600] [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: 12/23/2024] [Revised: 04/09/2025] [Accepted: 05/05/2025] [Indexed: 05/10/2025]
Abstract
Reineckea carnea (Andrews) Kunth (RCK) is known for its anti-inflammatory and antioxidant effects. But, its effects and underlying mechanisms on chronic obstructive pulmonary disease (COPD) are not well understood. This study aimed to evaluate the effects of RCK on COPD and to elucidate the mechanisms by which it modulates gut microbiota. A COPD mouse model was established through exposure to cigarette smoke (CS). Mice were then treated with oral administration of RCK aqueous extract. The anti-inflammatory effects and efficacy of RCK aqueous extract on COPD, as well as changes in microbiota composition, were evaluated. RCK aqueous extract ameliorated gut dysbiosis in CS-induced COPD mice by increasing the abundance of beneficial bacterial phyla and reducing the proliferation of pathogenic bacteria. Importantly, RCK treatment inhibited the expression of inflammatory mediators, such as IL-6, IL-8, and TNF-α at both mRNA levels and protein levels, attenuated oxidative stress in vivo in mice, and suppressed CS-induced activation of the NF-κB signaling pathway, thereby attenuating lung inflammation and restoring lung tissue structure. In conclusion, the beneficial effects of RCK aqueous extract on CS-induced COPD may be attributed to its anti-inflammatory and antioxidant properties as well as its ability to modulate gut microbial composition.
Collapse
Affiliation(s)
- Jiu-Qiong Chen
- Center for Tissue Engineering and Stem Cell Research,Guizhou Medical University, China; School of Pharmacy, Guizhou Medical University, China
| | - Xi-Jun Wu
- Jinyang Hospital Affiliated to Guizhou Medical University&The Second People's Hospital of Guiyang, China
| | - Xu-Xian Wu
- Center for Tissue Engineering and Stem Cell Research,Guizhou Medical University, China
| | - Bill D Geng
- School of Natural Science, University of Texas at Austin, Austin, TX 78712, USA
| | - Dan Zhou
- Center for Tissue Engineering and Stem Cell Research,Guizhou Medical University, China
| | - Jun Wen
- Department of Pharmacology, Xiamen Medical College, China
| | - Sze Chun Leo Chan
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Immunology Programme, The Life Science Institute, National University of Singapore, Singapore, Singapore
| | - Cen Jin
- Center for Tissue Engineering and Stem Cell Research,Guizhou Medical University, China
| | - Jian-Wei Xu
- Center for Tissue Engineering and Stem Cell Research,Guizhou Medical University, China; School of Pharmacy, Guizhou Medical University, China.
| | - Jun-Hou Lu
- Center for Tissue Engineering and Stem Cell Research,Guizhou Medical University, China.
| | - Guo Ge
- Department of Human Anatomy, School of Basic Medicine, Guizhou Medical University, China; Key Laboratory of Molecular Biology of Guizhou Medical University, China.
| |
Collapse
|
8
|
Ma J, Zhang J, Liu J, Zhao J, Wang X, Li Z, Lv T, Zhang Y. Ghrelin/GHSR system attenuates collagen-induced arthritis in mice and ameliorates inflammation in human rheumatoid arthritis fibroblast-like synoviocytes. Biochem Pharmacol 2025; 238:116973. [PMID: 40339721 DOI: 10.1016/j.bcp.2025.116973] [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: 12/27/2024] [Revised: 04/12/2025] [Accepted: 05/05/2025] [Indexed: 05/10/2025]
Abstract
Ghrelin, an acylated peptide hormone, acts through its sole known receptor, the growth hormone secretagogue receptor (GHSR).Previous research indicated that ghrelin may be involved in rheumatoid arthritis (RA), yet the specific mechanisms remain unclear. This study aimed to explore the mechanism of ghrelin in RA synovial inflammation. Serum and synovial tissue from RA patients were collected for ghrelin expression analysis. We conducted our study using a collagen-induced arthritis (CIA) mouse model and an in vitro model using fibroblast-like synoviocytes (FLSs) induced by tumor necrosis factor-alpha (TNF-α). RNA-sequencing was performed to identify the potential signaling pathways involved in RA. Ghsr shRNA interference was used to assess whether the ghrelin receptor was involved. Ghrelin expression was decreased in synovial tissue of RA patients, and was negatively associated with TNF-α in the synovial fluid. In vivo experiments, acyl-ghrelin effectively suppressed CIA development, and Ghsr-/- mice exhibited the significantly aggravated arthritis symptoms of CIA mice. RNA sequence analyses of synovial tissue in Ghsr-/- and wild type mice indicated that ghrelin/GHSRsystem may inhibit inflammation through the PI3K/AKT pathway. In RA-FLSs, we found that acyl-ghrelin significantly suppressed the TNF-α induced increase in p-PI3K, p-AKT, p-NF-κB p65, IL-6 and IL-1β in RA FLSs. The effects of acyl-ghrelin on inflammatory factors were attenuated by the PI3K/AKT agonists. Ghsr shRNA reversed the anti-inflammatory effects of acyl-ghrelin. These results indicated that ghrelin/GHSR system has an important role in RA and could be a suitable candidate for RA therapy.
Collapse
Affiliation(s)
- Junxian Ma
- Department of Rheumatology and Immunology, Tangdu Hospital, Air Force Medical University, 569 of Xinsi Road, Xi'an, Shaanxi, China; Department of Human Anatomy, Histology and Embryology, Air Force Medical University,169 of Changle Road, Xi'an, Shaanxi, China
| | - Jinshan Zhang
- Department of Human Anatomy, Histology and Embryology, Air Force Medical University,169 of Changle Road, Xi'an, Shaanxi, China; Department of Basic Medical Morphology, Medical College, Xijing University, 1 of Xijing Road, Xi'an, China
| | - Jie Liu
- Department of Rheumatology and Immunology, Tangdu Hospital, Air Force Medical University, 569 of Xinsi Road, Xi'an, Shaanxi, China
| | - Jie Zhao
- Department of Human Anatomy, Histology and Embryology, Air Force Medical University,169 of Changle Road, Xi'an, Shaanxi, China
| | - Xia Wang
- Department of Human Anatomy, Histology and Embryology, Air Force Medical University,169 of Changle Road, Xi'an, Shaanxi, China
| | - Zhen Li
- Department of Human Anatomy, Histology and Embryology, Air Force Medical University,169 of Changle Road, Xi'an, Shaanxi, China.
| | - Tingting Lv
- Department of Rheumatology and Immunology, Tangdu Hospital, Air Force Medical University, 569 of Xinsi Road, Xi'an, Shaanxi, China.
| | - Yan Zhang
- Department of Rheumatology and Immunology, Tangdu Hospital, Air Force Medical University, 569 of Xinsi Road, Xi'an, Shaanxi, China.
| |
Collapse
|
9
|
Caponio GR, Difonzo G, Troilo M, Marcotuli I, Gadaleta A, Tamma G, Gargano ML, Cirlincione F. Enhancing the Nutritional and Health-Related Properties of Taralli Through the Use of Pleurotus eryngii: Focus on Antioxidant and Anti-Inflammatory Properties. Antioxidants (Basel) 2025; 14:550. [PMID: 40427432 PMCID: PMC12108185 DOI: 10.3390/antiox14050550] [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: 04/03/2025] [Revised: 04/30/2025] [Accepted: 05/01/2025] [Indexed: 05/29/2025] Open
Abstract
In this study, a portion of whole durum wheat semolina was replaced with Pleurotus eryngii powder (PeP) at concentrations of 5% and 10% (w/w) to produce two taralli variants, TPE5 and TPE10. The impact of PeP on the technological, chemical, physical, and sensory properties of taralli was evaluated. The functional characteristics of the enriched taralli were assessed employing HCT8 human colon carcinoma cells as the experimental model. The inclusion of PeP in taralli increased total dietary fiber, meeting the "high fiber" criteria under Regulation 1924/2006 while also enhancing the total phenol content. The higher fiber and polyphenol content in the enriched samples contributed to a significant reduction in glycemic index and starch hydrolysis. Treatment with PeP-enriched taralli resulted in a notable decrease in intracellular ROS levels in HCT8 cells, demonstrating strong antioxidant potential. Furthermore, TPE5 exerted beneficial effects by reducing inflammation-evidenced by a significant decrease in NFkB phosphorylation at serine 536-and by promoting apoptosis. These effects are likely mediated through the regulation of intracellular oxidative states. Overall, these findings indicate that PeP enrichment enhances the nutritional profile of taralli and provides potential health benefits, reinforcing its role as a valuable functional ingredient.
Collapse
Affiliation(s)
- Giusy Rita Caponio
- Department of Bioscience, Biotechnology and Environment, University of Bari Aldo Moro, Via Orabona 4, 70125 Bari, Italy; (G.R.C.); (G.T.)
| | - Graziana Difonzo
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, Via Amendola 165/A, 70126 Bari, Italy; (M.T.); (I.M.); (A.G.); (M.L.G.); (F.C.)
| | - Marica Troilo
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, Via Amendola 165/A, 70126 Bari, Italy; (M.T.); (I.M.); (A.G.); (M.L.G.); (F.C.)
| | - Ilaria Marcotuli
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, Via Amendola 165/A, 70126 Bari, Italy; (M.T.); (I.M.); (A.G.); (M.L.G.); (F.C.)
| | - Agata Gadaleta
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, Via Amendola 165/A, 70126 Bari, Italy; (M.T.); (I.M.); (A.G.); (M.L.G.); (F.C.)
| | - Grazia Tamma
- Department of Bioscience, Biotechnology and Environment, University of Bari Aldo Moro, Via Orabona 4, 70125 Bari, Italy; (G.R.C.); (G.T.)
| | - Maria Letizia Gargano
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, Via Amendola 165/A, 70126 Bari, Italy; (M.T.); (I.M.); (A.G.); (M.L.G.); (F.C.)
| | - Fortunato Cirlincione
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, Via Amendola 165/A, 70126 Bari, Italy; (M.T.); (I.M.); (A.G.); (M.L.G.); (F.C.)
| |
Collapse
|
10
|
Alsaab J, Sarawi WS, Alhusaini AM, Hasan IH, Alturaif S, Ali RA, Alrasheed NM, Mohammad R, Algarzae NK. Procyanidin B2 mitigates methotrexate-induced hepatic pyroptosis by suppressing TLR4/NF-κB and caspase-3/GSDME pathways. Food Chem Toxicol 2025; 199:115341. [PMID: 39988050 DOI: 10.1016/j.fct.2025.115341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 02/08/2025] [Accepted: 02/19/2025] [Indexed: 02/25/2025]
Abstract
Methotrexate (MTX), a potent chemotherapeutic and immunosuppressive agent, is widely used for cancer and autoimmune diseases. MTX-induced hepatotoxicity is a well-recognized adverse response, even at relatively low doses. This study investigates the possible protective effects of procyanidin B2 (PCB2) on MTX-induced hepatotoxicity. Rats were orally treated with PCB2 (40 mg/kg) for 10 days, followed by a single intraperitoneal MTX injection (20 mg/kg) on day 8. The study also included a positive control group treated with quercetin (20 mg/kg), a known antioxidant, alongside MTX. The results revealed that MTX-induced hepatic injury was evidenced by elevation in serum transaminases. This elevation was accompanied by hepatic oxidative stress due to an imbalance in oxidative/antioxidant markers, specifically elevated malondialdehyde (MDA) and decreased glutathione (GSH) levels and superoxide dismutase (SOD) activity. The inflammatory cytokines, including tumor necrosis factor-alpha (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6), were markedly upregulated in the liver of MTX-intoxicated rats. Additionally, the expressions of nuclear factor kappa B (NF-κB), toll-like receptor 4 (TLR4), caspase-3 and gasdermin E (GSDME) were significantly increased in MTX rats. The use of PCB2 significantly ameliorated the deleterious effect of MTX on previous parameters by restoring oxidant/antioxidant balance, decreasing the inflammatory markers, and normalizing the expression of NF-κB, TLR4, caspase-3 and GSDME. In conclusion, this study uncovered the potential role of PCB2 on MTX-induced hepatotoxicity, confirming its antioxidant, anti-inflammatory, and anti-pyroptosis effects yet, further studies are needed to support its use as a protective therapy against such toxicity.
Collapse
Affiliation(s)
- Juman Alsaab
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 22452, Riyadh, 11495, Saudi Arabia.
| | - Wedad S Sarawi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 22452, Riyadh, 11495, Saudi Arabia.
| | - Ahlam M Alhusaini
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 22452, Riyadh, 11495, Saudi Arabia.
| | - Iman H Hasan
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 22452, Riyadh, 11495, Saudi Arabia.
| | - Sumayya Alturaif
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 22452, Riyadh, 11495, Saudi Arabia.
| | - Rehab A Ali
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 22452, Riyadh, 11495, Saudi Arabia.
| | - Nouf M Alrasheed
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 22452, Riyadh, 11495, Saudi Arabia.
| | - Raeesa Mohammad
- Department of Histology, College of Medicine, King Saud University, P.O. Box 2925, Riyadh, 11461, Saudi Arabia.
| | - Norah K Algarzae
- Department of Physiology, College of Medicine, King Saud University, P.O. Box 2925, Riyadh, 11461, Saudi Arabia.
| |
Collapse
|
11
|
Gong X, Xu L, Cai P. Friend or foe of tripartite motif-containing protein 21 in cardiovascular disease: A review. Int J Biol Macromol 2025; 308:142682. [PMID: 40164260 DOI: 10.1016/j.ijbiomac.2025.142682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 03/26/2025] [Accepted: 03/28/2025] [Indexed: 04/02/2025]
Abstract
As an E3 ubiquitin ligase and an Fc receptor, tripartite motif-containing protein 21 (TRIM21) plays a crucial role in immune defense, signal transduction, and cellular regulation. TRIM21 is widely expressed in various tissues, but it is particularly abundant in cardiovascular tissues and is involved in the pathogenesis of various cardiovascular diseases (CVDs). However, although TRIM21 is involved in the regulation of several key molecular pathways in the immune system, its specific role in CVD remains unclear. In this review, we comprehensively summarize the regulatory role of TRIM21 in signaling pathways and discuss the function of TRIM21 in CVD, to provide a systematic understanding of this important protein in CVD and offer insights for further research into the pathogenesis of CVD and its potential applications.
Collapse
Affiliation(s)
- Xiangmei Gong
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Xu
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pengcheng Cai
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
12
|
Yu C, Qiu G, Liu X, Xie Q, Lin Z, Wang F, Cai L. Anti-inflammatory effect and mechanism of stytontriterpene D on RAW264.7 cells and zebrafish. Front Pharmacol 2025; 16:1559022. [PMID: 40356999 PMCID: PMC12066662 DOI: 10.3389/fphar.2025.1559022] [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: 01/11/2025] [Accepted: 04/11/2025] [Indexed: 05/15/2025] Open
Abstract
Introduction Stytontriterpene D ( STD ) is a compound isolated from dried resin of Styrax tonkinensis (Pierre) Craib ex Hartw. In this study, we explored the anti-inflammatory effect of STD in vitro and in vivo and examined its potential anti-inflammatory mechanism for the first time. Methods In vitro, we evaluated the toxicity of STD to RAW 264.7 cells using the CCK8 method and detected the reactive oxygen species (ROS) and nitric oxide (NO) contents in cells using diacetyldichlorofluorescein (DCFH-DA) and the Griess method. We detected the levels of interleukin-6 (IL-6), interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-κ), inducible nitric oxide synthase (iNOS), interleukin-10 (IL-10), and arginase-1 (ARG1) via enzyme-linked immunosorbent assay and measured the expression of related proteins in the NF-αB pathway via western blotting. The toxicity of STD to AB zebrafish was detected in vivo, and the recruitment of neutrophils and macrophages was evaluated in tail cut -induced and copper sulfate -induced zebrafish inflammation models. We used quantitative real-time polymerase chain reaction to study the expression of inflammation-related genes in zebrafish with inflammation induced by copper sulfate. Results In lipopolysaccharide (LPS)-induced RAW 264.7 cells, STD decreased IL-6, IL-1β, NO, ROS, and TNF-α production, and increased the expression of IL-10 and ARG1 while also blocking inhibitory κBα (IκBα) phosphorylation and suppressing P65 nuclear translocation. STD also reduced the recruitment of inflammatory cells in zebrafish with inflammation induced by tail cutting and copper sulfate. STD not only reduced the copper sulfate-induced gene expression of zebrafish inflammatory factors, but it also inhibited the mRNA levels of NF-κB p65 and IκBα. Conclusion These results demonstrated that STD has an obvious anti-inflammatory effect, and its intrinsic molecular mechanism is possibly caused by inhibiting the NF-κB signaling pathway and regulating the phenotypic changes of M1 and M2 macrophages. Thus, STD may play a potential role in the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Chuqin Yu
- Centre for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Key Laboratory for Research and Evaluation of Pharmaceutical Preparations, Guangzhou, China
- Guangdong Engineering and Technology Research Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, China
| | - Gao Qiu
- Centre for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Key Laboratory for Research and Evaluation of Pharmaceutical Preparations, Guangzhou, China
- Guangdong Engineering and Technology Research Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xiangying Liu
- Centre for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Key Laboratory for Research and Evaluation of Pharmaceutical Preparations, Guangzhou, China
- Guangdong Engineering and Technology Research Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, China
| | - Quanwei Xie
- Centre for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Key Laboratory for Research and Evaluation of Pharmaceutical Preparations, Guangzhou, China
- Guangdong Engineering and Technology Research Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, China
| | - Zonghao Lin
- Centre for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Key Laboratory for Research and Evaluation of Pharmaceutical Preparations, Guangzhou, China
- Guangdong Engineering and Technology Research Center of Topical Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou, China
| | - Feng Wang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, China
| | - Lei Cai
- Guangdong Provincial Biotechnology Research Institute (Guangdong Provincial Laboratory Animals Monitoring Center), Guangzhou, China
| |
Collapse
|
13
|
He Z, Tan X, Yuan M, Chen L, Meng Y, Wang Q, Hu J, Qiu Z, Yang Y. Anethole trithione mitigates LPS/D-Gal-induced acute liver injury by suppressing ROS production and NF-κB activity. Int Immunopharmacol 2025; 152:114371. [PMID: 40054324 DOI: 10.1016/j.intimp.2025.114371] [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: 09/04/2024] [Revised: 01/14/2025] [Accepted: 02/24/2025] [Indexed: 03/24/2025]
Abstract
Acute liver injury (ALI) is a prevalent form of hepatic disease associated with significant morbidity and mortality due to medical treatments, exposure to toxins or viral infections. Anethole trithione (ATT) is a heterocyclic sulfur compound recognized for its chemoprotective properties against cancer and drug-induced toxicity. This study aimed to evaluate the effectiveness of ATT in the treatment of ALI. The therapeutic effects of ATT on hepatic injury were evaluated in vivo by inducing ALI in mice through the administration of lipopolysaccharide (LPS) and D-galactosamine (D-Gal). Additionally, HepG2 and Huh7 cells exposed to LPS were utilized to investigate the underlying mechanisms in vitro. The results indicated that ATT significantly reduced the production of reactive oxygen species (ROS), mitigated oxidative stress-related biochemical markers, and inhibited hepatocyte apoptosis in vivo, resulting in marked improvement in ALI in the murine model. Mechanistic studies conducted both in vivo and in vitro demonstrated that ATT alleviates LPS/D-Gal-induced ALI by inhibiting ROS production and the activity of nuclear factor-kappa B (NF-κB). Collectively, these findings underscore the potential therapeutic benefits of ATT in the management of ALI.
Collapse
Affiliation(s)
- Zhen He
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Xiangyun Tan
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Ming Yuan
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Liang Chen
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Yan Meng
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Qi Wang
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China
| | - Junjie Hu
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China.
| | - Zhenpeng Qiu
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan 430065, China; Center of Traditional Chinese Medicine Modernization for Liver Diseases, Hubei University of Chinese Medicine, Wuhan 430065, China; Hubei Key Laboratory of Resources and Chemistry of Chinese Medicine, Hubei University of Chinese Medicine, Wuhan 430065, China; Hubei Shizhen Laboratory, Wuhan 430061, China.
| | - Yuan Yang
- Institute of Maternal and Child Health, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430016, China.
| |
Collapse
|
14
|
Liu B, Yao Z, Song L, Sun C, Shen C, Cheng F, Cheng Z, Zhang R, Liu R. Vitexin alleviates lipid metabolism disorders and hepatic injury in obese mice through the PI3K/AKT/mTOR/SREBP-1c pathway. Eur J Med Chem 2025; 287:117379. [PMID: 39947052 DOI: 10.1016/j.ejmech.2025.117379] [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: 02/02/2025] [Accepted: 02/06/2025] [Indexed: 02/24/2025]
Abstract
Obesity is recognized as a metabolic disorder, and its treatment and management pose ongoing challenges worldwide. Hawthorn, a traditional Chinese herb used to alleviate digestive issues and reduce blood lipid levels, has unclear mechanisms of action regarding its active components in the treatment of obesity. This study investigated the anti-obesity effects of vitexin, a major flavonoid compound found in hawthorn, in high-fat diet (HFD)-induced C57BL/6 mice. The results demonstrated that vitexin significantly reduced body weight, liver weight, blood lipid levels, and inflammatory markers in obese mice, while also inhibiting hepatic lipid accumulation. Mechanistic studies revealed that vitexin likely suppresses adipogenesis by modulating the PI3K-AKT signaling pathway, as evidenced by reduced expression of PI3K, phosphorylated AKT, phosphorylated mTOR, and SREBP-1c in the livers of vitexin-treated obese mice. Additionally, vitexin inhibited NFκB expression by regulating IκBα phosphorylation, thereby alleviating obesity-induced liver injury. These findings suggest that vitexin may be the primary active component in hawthorn responsible for reducing blood lipid levels, highlighting its potential in the treatment of obesity and its associated metabolic disorders.
Collapse
Affiliation(s)
- Bo Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Ziqing Yao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Lin Song
- Department of Pharmacy, Children' S Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, China
| | - Chen Sun
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Changhong Shen
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Fang Cheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Zefang Cheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Ruoqi Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Rong Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
15
|
Xue C, Chu Q, Shi Q, Zeng Y, Lu J, Li L. Wnt signaling pathways in biology and disease: mechanisms and therapeutic advances. Signal Transduct Target Ther 2025; 10:106. [PMID: 40180907 PMCID: PMC11968978 DOI: 10.1038/s41392-025-02142-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/13/2024] [Accepted: 12/29/2024] [Indexed: 04/05/2025] Open
Abstract
The Wnt signaling pathway is critically involved in orchestrating cellular functions such as proliferation, migration, survival, and cell fate determination during development. Given its pivotal role in cellular communication, aberrant Wnt signaling has been extensively linked to the pathogenesis of various diseases. This review offers an in-depth analysis of the Wnt pathway, detailing its signal transduction mechanisms and principal components. Furthermore, the complex network of interactions between Wnt cascades and other key signaling pathways, such as Notch, Hedgehog, TGF-β, FGF, and NF-κB, is explored. Genetic mutations affecting the Wnt pathway play a pivotal role in disease progression, with particular emphasis on Wnt signaling's involvement in cancer stem cell biology and the tumor microenvironment. Additionally, this review underscores the diverse mechanisms through which Wnt signaling contributes to diseases such as cardiovascular conditions, neurodegenerative disorders, metabolic syndromes, autoimmune diseases, and cancer. Finally, a comprehensive overview of the therapeutic progress targeting Wnt signaling was given, and the latest progress in disease treatment targeting key components of the Wnt signaling pathway was summarized in detail, including Wnt ligands/receptors, β-catenin destruction complexes, and β-catenin/TCF transcription complexes. The development of small molecule inhibitors, monoclonal antibodies, and combination therapy strategies was emphasized, while the current potential therapeutic challenges were summarized. This aims to enhance the current understanding of this key pathway.
Collapse
Affiliation(s)
- Chen Xue
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qingfei Chu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qingmiao Shi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yifan Zeng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Juan Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
16
|
Yeshna, Singh M, Monika, Kumar A, Garg V, Jhawat V. Pathophysiology and emerging therapeutic strategies for cervical spondylosis: The role of pro-inflammatory mediators, kinase inhibitors, and Organogel based drug delivery systems. Int Immunopharmacol 2025; 151:114350. [PMID: 40010157 DOI: 10.1016/j.intimp.2025.114350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/20/2025] [Accepted: 02/20/2025] [Indexed: 02/28/2025]
Abstract
Cervical spondylosis is a prevalent ailment characterized by chronic wear and degenerative changes affecting the cervical spine, leading to various clinical syndromes such as axial neck pain, cervical myelopathy, and cervical radiculopathy. The pathophysiology of the development of cervical alterations is multifaceted, with alterations in the normal physiology and pathogenesis of intervertebral disc degeneration. The involvement of pro-inflammatory mediators, such as interleukin-1, tumor necrosis factor-α, interleukin-4, interleukin-6, and interleukin-10, in the pathological processes associated with intervertebral disc degeneration offers potential therapeutic targets. The review also introduces kinase inhibitors as potential treatments for cervical spondylosis. Protein kinase inhibitors, including mitogen-activated protein kinase (MAPK), Janus kinase (JAK), and spleen tyrosine kinase (SYK), are explored for their anti-inflammatory properties. The article discusses their potential in modulating inflammatory signaling cascades and presents them as attractive candidates for treating immune-mediated disorders. Inhibitors of Nuclear Factor-κB, p38 MAPK, Jun-N terminal kinase (JNK), and Extracellular signal-regulated kinase (ERK) have shown efficacy in suppressing inflammatory responses, offering potential avenues for intervention in this prevalent condition. Organogels are semi-solid materials formed by trapping an organic solvent within a three-dimensional cross-linked network. They hold considerable potential in drug delivery, especially in enhancing drug solubility, facilitating controlled release, and improving skin penetration. These properties of organogels can help treat or alleviate the symptoms of cervical spondylosis.
Collapse
Affiliation(s)
- Yeshna
- Department of Pharmaceutical Science, School of Healthcare and Allied Science, GD Goenka University, Gurugram, Haryana, India
| | - Monika Singh
- Department of Pharmaceutical Science, School of Healthcare and Allied Science, GD Goenka University, Gurugram, Haryana, India
| | - Monika
- Department of Pharmaceutical Science, School of Healthcare and Allied Science, GD Goenka University, Gurugram, Haryana, India
| | - Ashok Kumar
- Faculty of Pharmacy, Kalinga University, Naya Raipur, Chhattisgarh, India
| | - Vandana Garg
- Department of Pharmaceutical Science, MD University, Rohtak, India
| | - Vikas Jhawat
- Department of Pharmaceutical Science, School of Healthcare and Allied Science, GD Goenka University, Gurugram, Haryana, India.
| |
Collapse
|
17
|
Kelly DM, Kelleher EM, Rothwell PM. The Kidney-Immune-Brain Axis: The Role of Inflammation in the Pathogenesis and Treatment of Stroke in Chronic Kidney Disease. Stroke 2025; 56:1069-1081. [PMID: 39851054 PMCID: PMC11932449 DOI: 10.1161/strokeaha.124.047070] [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] [Indexed: 01/25/2025]
Abstract
Cardiovascular diseases such as stroke are a major cause of morbidity and mortality for patients with chronic kidney disease (CKD). The underlying mechanisms connecting CKD and cardiovascular disease are yet to be fully elucidated, but inflammation is proposed to play an important role based on genetic association studies, studies of inflammatory biomarkers, and clinical trials of anti-inflammatory drug targets. There are multiple sources of both endogenous and exogenous inflammation in CKD, including increased production and decreased clearance of proinflammatory cytokines, oxidative stress, metabolic acidosis, chronic and recurrent infections, dialysis access, changes in adipose tissue metabolism, and disruptions in intestinal microbiota. This review focuses on the mechanisms of inflammation in CKD, dialysis and associated therapies, its proposed impact on stroke pathogenesis and prognosis, and the potential role of anti-inflammatory agents in the prevention and treatment of stroke in patients with CKD.
Collapse
Affiliation(s)
- Dearbhla M. Kelly
- Wolfson Centre for the Prevention of Stroke and Dementia, Nuffield Department of Clinical Neurosciences (D.M.K., P.M.R.)
| | - Eoin M. Kelleher
- Nuffield Department of Clinical Neurosciences (E.M.K.), University of Oxford, United Kingdom
| | - Peter M. Rothwell
- Wolfson Centre for the Prevention of Stroke and Dementia, Nuffield Department of Clinical Neurosciences (D.M.K., P.M.R.)
| |
Collapse
|
18
|
Hassan EA, Abdelnaser A, Ibrahim S, Yousef EH, Mosallam AM, Zayed SE. 5H Pyrolo(3,4-b)Pyrazin-5,7-(6H)-dione 6-(N-Chitosanimide nanoparticle) composite nano silver and encapsulation in γ-cyclodextrin: Synthesis, molecular docking, and biological evaluation for thyroid cancer treatment. Int J Biol Macromol 2025; 304:140859. [PMID: 39947539 DOI: 10.1016/j.ijbiomac.2025.140859] [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: 01/14/2025] [Accepted: 02/08/2025] [Indexed: 02/20/2025]
Abstract
BACKGROUND Thyroid cancer is rapidly increasing worldwide, with some patients facing poor prognosis and recurrence despite current treatments. Chitosan-based nanoparticles have exhibited exciting antitumor efficacy both in vitro and in vivo, which indicates that there is vast scope of clinical application. This study develops a anhydride-modified chitosan and anhydride-modified chitosan‑silver nanoparticles, encapsulated in γ-cyclodextrin to help drug delivery by safe way and enhance thyroid cancer therapy. METHODS 5H pyrolo(3,4-b)pyrazin-5,7-(6H)-dione-6-(N-chitosanimide nanoparticle(composite constructed with nano silver (B1) was prepared and the optimized formula was further investigated regarding FT-IR, X-RD, SEM and TEM. Furthermore, it was encapsulated in γ-CD, and an in vivo study was conducted to investigate its anticancer activity. The binding affinities of 2,3-Pyrazinedicarboxylic anhydride to inhibitor of kappa B kinase beta (IKK-β) was demonstrated by molecular docking. RESULTS SEM and TEM revealed that Ag NPs were mostly uniformly incorporated into the 5H pyrolo(3,4-b)pyrazin-5,7-(6H)-dione 6-(N-chitosanimide nanoparticle, while FT-IR and X-RD findings verified the formation of 5H pyrolo(3,4-b)pyrazin-5,7-(6H)-dione-6-(N-chitosanimide nanoparticle)/composite constructed with nano silver and encapsulated in γ-CD (B2). γ-CD encapsulation induced a significant enhancement in pyrazine thyroid antitumor activity in xenografic model. CONCLUSION B2 could be considered a promising formula for suppression of thyroid cancer by modulating NF-κB signaling pathway, and hence, future studies could be planned to transfer our formula to the clinical field.
Collapse
Affiliation(s)
- Entesar A Hassan
- Department of Chemistry, Faculty of Science, South Valley University, Qena, 83523, Egypt
| | - Amira Abdelnaser
- Department of Chemistry, Faculty of Science, South Valley University, Qena, 83523, Egypt
| | - Samar Ibrahim
- Department of Clinical Pharmacy & Pharmacy Practice, Faculty of Pharmacy, Galala University, Ataka, Egypt
| | - Eman H Yousef
- Pharmacology and Biochemistry Department, Faculty of Pharmacy, Horus University-Egypt, New Damietta 34518, Egypt.
| | - Ahmed M Mosallam
- Department of Chemistry, Faculty of Science, South Valley University, Qena, 83523, Egypt
| | - Salem E Zayed
- Department of Chemistry, Faculty of Science, South Valley University, Qena, 83523, Egypt
| |
Collapse
|
19
|
Hong Y, Wang D, Qian H, Jiang X, Wang Y, Liang X, Gao S, Hua C. Exploring the molecular mechanism of Tripterygium Wilfordii Hook F in treating systemic lupus erythematosus via network pharmacology and molecular docking. Clin Rheumatol 2025; 44:1549-1569. [PMID: 39934490 DOI: 10.1007/s10067-025-07311-4] [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: 05/11/2024] [Revised: 12/20/2024] [Accepted: 12/30/2024] [Indexed: 02/13/2025]
Abstract
BACKGROUND Tripterygium wilfordii Hook F (TwHF) is a prominent Chinese herbal formula. It exhibits significant clinical efficacy in treating systemic lupus erythematosus (SLE), though its mechanisms remain unclear. Our study employs network pharmacology and molecular docking to explore active compounds of TwHF and their associated targets for SLE treatment. METHODS Primary active compounds of TwHF and their targets were sourced from the TCMSP, SwissTargetPrediction, and UniProt databases. SLE-relevant target proteins were identified from the OMIM and GeneCards databases. Enrichment analyses were conducted to reveal results of common TwHF-SLE targets. STRING and Cytoscape software were used to systematically analyze and construct protein-protein interaction (PPI) networks, compound-target-pathway, and target-organ networks. Molecular docking was utilized to confirm the binding of key targets to the top active compounds. RESULTS A total of 14 active compounds and 300 overlapping targets between TwHF and SLE were identified. PPI network analysis revealed 29 core targets. Several pathways were found to contribute to the potential therapeutic effects of TwHF in SLE, including PI3K-Akt signaling pathway, Th17 cell differentiation, chemokine signaling, and T cell receptor signaling. Disease Ontology (DO) analysis highlighted the involvement of TwHF in genes associated with myocardial infarction (MI), atherosclerosis (AS), breast carcinoma, and ischemia. Molecular docking results demonstrated strong binding affinities, with 37 signal molecule-receptor interactions in SLE and 97 interactions in SLE-related MI and AS showing binding energies lower than -7 kJ/mol. CONCLUSIONS This research effectively anticipates the potent constituents, probable targets, and pathways implicated in treating SLE with TwHF, specifically addressing complications such as MI and AS. Comprehending the precise molecular mechanism targeting SLE of TwHF and its efficacious bioactive components furnishes a theoretical groundwork for enhancing its clinical utilization. Key Points •SLE is characterized by aberrant immune activation and persistent inflammation. •TwHF exerts immunomodulatory and anti-inflammatory effects. •TwHF exhibits prospects in the treatment of SLE with unknown molecular mechanisms. •Network pharmacology and molecular docking reveal promise in the mechanism of TwHF.
Collapse
Affiliation(s)
- Yanggang Hong
- The Second School of Medicine, Wenzhou Medical University, Zhejiang Province, Wenzhou, 325035, China
| | - Deqi Wang
- The First School of Medicine, Wenzhou Medical University, Zhejiang Province, Wenzhou, 325000, China
| | - Hengrong Qian
- The Second School of Medicine, Wenzhou Medical University, Zhejiang Province, Wenzhou, 325035, China
| | - Xiaoyang Jiang
- School of Ophthalmology & Optometry, School of Biomedical Engineering, Wenzhou Medical University, Zhejiang Province, Wenzhou, 325035, China
| | - Yi Wang
- The First School of Medicine, Wenzhou Medical University, Zhejiang Province, Wenzhou, 325000, China
| | - Xinyue Liang
- School of Ophthalmology & Optometry, School of Biomedical Engineering, Wenzhou Medical University, Zhejiang Province, Wenzhou, 325035, China
| | - Sheng Gao
- Laboratory Animal Center, Wenzhou Medical University, Zhejiang Province, Wenzhou, 325035, China.
| | - Chunyan Hua
- School of Basic Medical Sciences, Wenzhou Medical University, Zhejiang Province, Wenzhou, 325035, China.
| |
Collapse
|
20
|
You H, Zhang S, Zhang Y, Chen Q, Wu Y, Zhou Z, Zhao Z, Su B, Li X, Guo Y, Chen Y, Tang W, Liu B, Fan H, Geng S, Fang M, Li F, Liu G, Jiang C, Sun T. Engineered Bacterial Outer Membrane Vesicles-Based Doxorubicin and CD47-siRNA Co-Delivery Nanoplatform Overcomes Immune Resistance to Potentiate the Immunotherapy of Glioblastoma. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2418053. [PMID: 40035513 DOI: 10.1002/adma.202418053] [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: 11/20/2024] [Revised: 02/23/2025] [Indexed: 03/05/2025]
Abstract
Apart from the blood-brain barrier (BBB), the efficacy of immunotherapy for glioblastoma (GBM) is limited by the presence of intrinsic and adaptive immune resistance, implying that co-delivery of various immunotherapeutic agents or simultaneous regulation of different cells is urgently needed. Bacterial outer membrane vesicles (OMVs) offer a unique advantage in the treatment of GBM, owing to their multifunctional properties as carriers and immune adjuvants and their ability to cross the BBB. However, traditional OMVs can lead to toxic side effects and disruption of tight junctions in the BBB. Therefore, to enhance the in vivo safety and targeting capability of OMVs, we introduced engineered OMVs to reduce toxicity and further constructed a modularly assembled nanoplatform by performing simple peptide modifications. This nanoplatform demonstrates satisfactory biosafety and is able to continuously cross the BBB and target GBM with the assistance of Angiopep-2. Subsequently, immunogenic substances on OMVs, along with carried small-interfering RNA (siRNA) and doxorubicin, can promote and enhance the reprogramming and phagocytic abilities of macrophages and microglia, respectively, and increase the immunogenicity of GBM, ultimately overcoming GBM immune resistance to enhance the efficacy of immunotherapy. This OMVs-based nanoplatform provides a new paradigm and insights into the development of immunotherapy for GBM.
Collapse
Affiliation(s)
- Haoyu You
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Shilin Zhang
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Yiwen Zhang
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Qinjun Chen
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Yuxing Wu
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Zheng Zhou
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Zhenhao Zhao
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Boyu Su
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Xuwen Li
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Yun Guo
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Yun Chen
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Weiyi Tang
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Bing Liu
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Hongrui Fan
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Shuo Geng
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Mingzhu Fang
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Fangxin Li
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Guangna Liu
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Chen Jiang
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, 201203, China
- Department of Digestive Diseases, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Tao Sun
- Key Laboratory of Smart Drug Delivery/Innovative Center for New Drug Development of Immune Inflammatory Diseases (Ministry of Education), Minhang Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, 201203, China
- Quzhou Fudan Institute, Quzhou, 324003, China
| |
Collapse
|
21
|
Luo S, Wu X, Wang H, Zhang Y, Xie L. Nitrate induced hepatic fibrosis in tadpoles of Bufo gargarizans by mediating alterations in toll-like receptor signaling pathways. ENVIRONMENTAL RESEARCH 2025; 270:120961. [PMID: 39875068 DOI: 10.1016/j.envres.2025.120961] [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: 11/18/2024] [Revised: 01/22/2025] [Accepted: 01/25/2025] [Indexed: 01/30/2025]
Abstract
The nitrate pollution has become an increasingly serious environmental problem worldwide, and the toxic effects of elevated nitrate levels in the environment on aquatic animals remain to be elucidated. The purpose of the present study was to investigate the mechanisms of liver injury to tadpoles after exposure to nitrate from embryonic to metamorphic climax and to assess the recovery process of liver function after cessation of exposure. In the group with continuous nitrate exposure, the livers and thyroid of tadpoles showed remarkably histological lesions, of this with structural disorganization of the hepatocytes, cellular atrophy, and fibrosis, as well as significant reduction in the follicular and colloidal area of the thyroid. Meanwhile, the expression levels of genes related to inflammatory signaling pathways, such as TLR2, TLR6 and NF-κB, were significant elevated. After termination of exposure at Gs23, liver damage (histologic, ultrastructural, and molecular levels) was almost completely recovered, whereas thyroid gland damage was irreversible. Overall, this study shed light on the harmful effects of nitrate pollution on amphibian health and emphasizes the importance of controlling nitrate emissions in the environment.
Collapse
Affiliation(s)
- Shuangyan Luo
- College of Life and Environmental Science, Wenzhou University, 325003, Wenzhou, China; College of Life Science, Shaanxi Normal University, 710119, Xi'an, China
| | - Xueyi Wu
- College of Life and Environmental Science, Wenzhou University, 325003, Wenzhou, China
| | - Hongyuan Wang
- College of Life Science, Shaanxi Normal University, 710119, Xi'an, China
| | - Yongpu Zhang
- College of Life and Environmental Science, Wenzhou University, 325003, Wenzhou, China; Zhejiang Provincial Key Laboratory for Subtropical Water Environment and Marine Biological Resources Protection, Wenzhou University, 325003, Wenzhou, China.
| | - Lei Xie
- College of Life and Environmental Science, Wenzhou University, 325003, Wenzhou, China; Zhejiang Provincial Key Laboratory for Subtropical Water Environment and Marine Biological Resources Protection, Wenzhou University, 325003, Wenzhou, China.
| |
Collapse
|
22
|
Deng Y, Zhong G, Jin T, Wang J, Peng X, Zhou K, Chen X, Cao X. Mechanism exploration of Wenshen Jianpi Decoction on renoprotection in diabetic nephropathy via transcriptomics and metabolomics. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156446. [PMID: 39914065 DOI: 10.1016/j.phymed.2025.156446] [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: 09/23/2024] [Revised: 01/05/2025] [Accepted: 01/28/2025] [Indexed: 05/07/2025]
Abstract
BACKGROUND Diabetic nephropathy (DN) is a severe chronic microvascular complications of diabetes mellitus and the leading cause of end-stage renal disease. Although many therapeutic approaches have been developed for treatment of DN, there is still a risk of disease progression. Wenshen Jianpi Decoction (WSJPD), a traditional Chinese medicine, have demonstrated considerable clinical efficacy in treating DN. However, the therapeutic mechanisms of WSJPD remain unclear. PURPOSE The study aimed to assess the potential anti-renal injury effects of WSJPD as well as the underlying mechanism. METHODS Network pharmacology was performed to predict the potential targets of WSJPD in DN. Subsequently, HE, PAS, Masson staining, TEM and ELISA were used to assess the effects of WSJPD on renal injury in DN rats. Transcriptomics, metabolomics, and qRT-PCR were employed to analyze the potential mechanism. RESULTS Network pharmacology analysis revealed that WSJPD might exert a beneficial impact on renal inflammation via the AGE-RAGE signaling pathway. The results of animal experiments indicated that WSJPD had a renoprotective effect by renal dysfunction improvement, inflammation inhibition, glycogen accumulation and podocyte injury suppression.Transcriptomic measures discovered that 212 genes were up-regulated in the DN model group and down-regulated in the WSJPD group. Conversely, 80 genes were down-regulated in the DN model group and up-regulated in the WSJPD group. Moreover, 14 metabolites from rat serum samples were identified as candidate biomarkers. Further analysis demonstrated that WSJPD ameliorated the metabolic disorders and exerted a protective effect by up-regulation of uridine-mediated pyrimidine metabolism, inhibition of inflammation mediated by the NF-κB and TNF signaling pathways, and inhibition of oxidative stress and inflammation mediated by the AGE-RAGE signaling pathway. CONCLUSION Our findings showed that WSJPD ameliorated renal damage in DN model rats by regulating kidney pyrimidine metabolism, inflammation-related pathways, and AGE-RAGE signaling pathway. The potential renoprotective effects were verified by histopathology, ELISA, and qRT-PCR. This study not only support a foundation for interpretation of the therapeutic effects of WSJPD on renal lesions, but also provide data support for further explorations of the novel compound preparations to improve renal function in DN.
Collapse
Affiliation(s)
- Yingping Deng
- Ningbo Municipal Hospital of Traditional Chinese Medicine, Affiliated to Zhejiang Chinese Medical University, Ningbo 315000, Zhejiang, PR China
| | - Guanghui Zhong
- Ningbo Municipal Hospital of Traditional Chinese Medicine, Affiliated to Zhejiang Chinese Medical University, Ningbo 315000, Zhejiang, PR China
| | - Tinglong Jin
- Ningbo Municipal Hospital of Traditional Chinese Medicine, Affiliated to Zhejiang Chinese Medical University, Ningbo 315000, Zhejiang, PR China
| | - Jin Wang
- Ningbo Municipal Hospital of Traditional Chinese Medicine, Affiliated to Zhejiang Chinese Medical University, Ningbo 315000, Zhejiang, PR China
| | - Xin Peng
- Ningbo Municipal Hospital of Traditional Chinese Medicine, Affiliated to Zhejiang Chinese Medical University, Ningbo 315000, Zhejiang, PR China
| | - Kai Zhou
- Ningbo Municipal Hospital of Traditional Chinese Medicine, Affiliated to Zhejiang Chinese Medical University, Ningbo 315000, Zhejiang, PR China
| | - Xiabo Chen
- Ningbo Municipal Hospital of Traditional Chinese Medicine, Affiliated to Zhejiang Chinese Medical University, Ningbo 315000, Zhejiang, PR China.
| | - Xiaodan Cao
- Ningbo Municipal Hospital of Traditional Chinese Medicine, Affiliated to Zhejiang Chinese Medical University, Ningbo 315000, Zhejiang, PR China.
| |
Collapse
|
23
|
Zhang RY, Wang ZX, Zhang MY, Wang YF, Zhou SL, Xu JL, Lin WX, Ji TR, Chen YD, Lu T, Li NG, Shi ZH. MALT1 Inhibitors and Degraders: Strategies for NF-κB-Driven Malignancies. J Med Chem 2025; 68:5075-5096. [PMID: 39999563 DOI: 10.1021/acs.jmedchem.4c02873] [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: 02/27/2025]
Abstract
Mucosa-associated lymphoid tissue protein 1 (MALT1), a cysteine protease and the sole paracaspase in humans, plays a pivotal role in the survival and proliferation of NF-κB-dependent malignant cancers, particularly MALT lymphoma and diffuse large B-cell lymphoma (DLBCL). Dysregulated MALT1 activity is implicated in various malignancies, highlighting its importance as a therapeutic target. This Perspective provides an overview of MALT1's structural and functional characteristics, summarizes recent advancements in small-molecule inhibitors and degraders targeting this protein, and discusses compound structures, structure-activity relationship (SAR) analyses, and biological activities. We aim to inform future research efforts to enhance the activity, selectivity, and pharmacological properties of MALT1-targeting compounds, establishing a foundational framework for drug development in this critical area of cancer therapy.
Collapse
Affiliation(s)
- Ru-Yue Zhang
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, China
| | - Zi-Xuan Wang
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Meng-Yuan Zhang
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Yu-Fan Wang
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, China
| | - Si-Li Zhou
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, China
| | - Jia-Lu Xu
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, China
| | - Wen-Xuan Lin
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, China
| | - Tian-Rui Ji
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, China
| | - Ya-Dong Chen
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, China
| | - Tao Lu
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, China
| | - Nian-Guang Li
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Zhi-Hao Shi
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, China
| |
Collapse
|
24
|
Cao J, Wang Y, Lin Q, Wang S, Shen Y, Zhang L, Li W, Chen L, Liu C, Yao S, Shuai L, Chen X, Li Z, Chang Y. IL-1β stimulates ADAMTS9 expression and contributes to preterm prelabor rupture of membranes. Cell Commun Signal 2025; 23:127. [PMID: 40057799 PMCID: PMC11890524 DOI: 10.1186/s12964-025-02120-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 02/22/2025] [Indexed: 05/13/2025] Open
Abstract
BACKGROUND Preterm prelabor rupture of membranes (pPROM) is a leading cause of neonatal morbidity and mortality. While intra-amniotic infection is a well-established driver of pPROM, the role of sterile intra-amniotic inflammation remains unclear. Recent evidence suggests that interleukin-1 beta (IL-1β) promotes extracellular matrix (ECM) remodeling via downstream effectors, a disintegrin-like and metalloproteinase domain with thrombospondin type 1 motif 9 (ADAMTS9), while protein O-fucosyltransferase 2 (POFUT2) facilitates its O-fucosylation and secretion, amplifying ECM degradation. This study investigates how IL-1β-triggered nuclear factor kappa-B (NF-κB) activation promotes ADAMTS9 and POFUT2 expression, ultimately driving fetal membrane ECM remodeling and weakening in pPROM without signs of intra-amniotic infection. METHODS A nested case-control study included maternal serum and fetal membrane samples from 60 pregnant women (34 pPROM, 26 full-term births [FTB]). ELISA measured serum levels of IL-1β and ADAMTS9, and their correlations were analyzed. Mechanistic studies utilized primary human amniotic epithelial cells (hAECs) and fetal membrane-decidua explants with IL-1β treatment. The role of NF-κB was explored using chromatin immunoprecipitation (ChIP) and luciferase assays to assess NF-κB binding to the promoters of ADAMTS9 and POFUT2. A murine model of sterile intra-amniotic inflammation under ultrasound-guided IL-1β injection was used to validate in vitro findings and assess pregnancy outcomes. RESULTS Serum IL-1β and ADAMTS9 levels at 16 weeks of gestation were significantly higher in pPROM cases compared to FTB controls (P < 0.001). A combined model of these biomarkers demonstrated high predictive accuracy for pPROM (AUC = 0.83). Mechanistically, IL-1β activated NF-κB, leading to its binding to the promoters of ADAMTS9 and POFUT2. NF-κB activation promoted ADAMTS9 expression, while POFUT2 enhanced its secretion. Together, these processes drove versican degradation and ECM weakening. Intra-amniotic administration of IL-1β in mice induced fetal membrane weakening, preterm birth, and adverse neonatal outcomes, which were mitigated by the NF-κB inhibitor BAY 11-7082 treatment. CONCLUSION Maternal serum ADAMTS9 levels at mid-gestation are promising non-invasive biomarkers for pPROM risk stratification. Mechanistically, IL-1β-induced NF-κB activation promotes ADAMTS9 expression and POFUT2-dependent secretion, contributing to fetal membrane weakening. These findings provide new insights into the role and potential therapeutic target for sterile intra-amniotic inflammation in pPROM.
Collapse
Affiliation(s)
- Jiasong Cao
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, 300100, China
- Nankai University Affiliated Hospital of Obstetrics and Gynecology, Tianjin, 300100, China
- Tianjin Institute of Gynecology Obstetrics, Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, 300100, China
| | - Yixin Wang
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Qimei Lin
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, 300100, China
- Nankai University Affiliated Hospital of Obstetrics and Gynecology, Tianjin, 300100, China
- Tianjin Institute of Gynecology Obstetrics, Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, 300100, China
| | - Shuqi Wang
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, 300100, China
- Nankai University Affiliated Hospital of Obstetrics and Gynecology, Tianjin, 300100, China
| | - Yongmei Shen
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, 300100, China
- Nankai University Affiliated Hospital of Obstetrics and Gynecology, Tianjin, 300100, China
- Tianjin Institute of Gynecology Obstetrics, Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, 300100, China
| | - Lei Zhang
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, 300100, China
- Nankai University Affiliated Hospital of Obstetrics and Gynecology, Tianjin, 300100, China
| | - Wen Li
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, 300100, China
- Nankai University Affiliated Hospital of Obstetrics and Gynecology, Tianjin, 300100, China
- Tianjin Institute of Gynecology Obstetrics, Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, 300100, China
| | - Ling Chen
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, 300100, China
- Nankai University Affiliated Hospital of Obstetrics and Gynecology, Tianjin, 300100, China
| | - Chunliu Liu
- Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, 300120, China
| | - Shihan Yao
- Nankai University Affiliated Hospital of Obstetrics and Gynecology, Tianjin, 300100, China
| | - Ling Shuai
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, 300100, China
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin, 300350, China
| | - Xu Chen
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, 300100, China
- Nankai University Affiliated Hospital of Obstetrics and Gynecology, Tianjin, 300100, China
| | - Zongjin Li
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, 300100, China.
- School of Medicine, Nankai University, Tianjin, 300071, China.
- Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin, 300071, China.
| | - Ying Chang
- Tianjin Key Laboratory of Human Development and Reproductive Regulation, Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, 300100, China.
- Nankai University Affiliated Hospital of Obstetrics and Gynecology, Tianjin, 300100, China.
- Medical School, Tianjin University, Tianjin, 300072, China.
| |
Collapse
|
25
|
Huang Y, Yang J, Lu T, Shao C, Wan H. Puerarin Alleviates Cerebral Ischemia-Reperfusion Injury by Inhibiting Ferroptosis Through SLC7A11/GPX4/ACSL4 Axis and Alleviate Pyroptosis Through Caspase-1/GSDMD Axis. Mol Neurobiol 2025:10.1007/s12035-025-04798-5. [PMID: 40056342 DOI: 10.1007/s12035-025-04798-5] [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: 11/16/2024] [Accepted: 02/21/2025] [Indexed: 03/10/2025]
Abstract
Cerebral ischemia-reperfusion (CIRI) represents a complex disease entity that encompasses multiple pathways. The occurrence of CIRI induces cerebral infarction, accompanied by brain tissue necrosis and focal neuronal impairment. Previous studies have demonstrated that ferroptosis, a specific cell death pathway implicated in CIRI, plays a crucial role in mediating the pathophysiological process of this condition. Puerarin, is known to possess vasodilatory, antioxidant, and neuroprotective properties. However, its precise role in ferroptosis as well as the underlying mechanisms remains elusive. In this study, we delved into the neuroprotective mechanisms of puerarin using both the rat middle cerebral artery occlusion (MCAO) model and the HT22 cell model of oxygen-glucose deprivation/reperfusion (OGD/R). In the MCAO model, puerarin was found to exhibit an inhibitory effect on ACSL4, which was consistent with that of rosiglitazone. Simultaneously, it was capable of counteracting the inhibition of GPX4 by RSL3. These findings suggest that puerarin modulates GPX4 and ACSL4, thereby exerting an inhibitory effect on ferroptosis. The ferroptosis-protective effect of puerarin was further corroborated in the OGD/R through a positive control experiment with ferrostatin-1, a lipid peroxidation inhibitor. Furthermore, we also recognized the importance of other cell death modalities, such as pyroptosis. Consequently, we verified the neuroprotective effect of puerarin by examining the influence of caspase-1 and GSDMD in HT22. Mechanistically, puerarin alleviates CIRI by respectively inhibiting ferroptosis through the SLC7A11/GPX4/ACSL4 axis and pyroptosis through the caspase-1/GSDMD axis. This research provides novel insights into the targeting and therapeutic potential of puerarin for the treatment of CIRI.
Collapse
Affiliation(s)
- Ying Huang
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Jiehong Yang
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
- Key Laboratory of TCM Encephalopathy of Zhejiang Province, Hangzhou, 310053, Zhejiang, China
| | - Ting Lu
- College of Life Science, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China
| | - Chongyu Shao
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
- Key Laboratory of TCM Encephalopathy of Zhejiang Province, Hangzhou, 310053, Zhejiang, China.
| | - Haitong Wan
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, Zhejiang, China.
- Key Laboratory of TCM Encephalopathy of Zhejiang Province, Hangzhou, 310053, Zhejiang, China.
| |
Collapse
|
26
|
Mahjoubin-Tehran M, Rezaei S, Butler AE, Sahebkar A. Decoy oligonucleotides targeting NF-κB: a promising therapeutic approach for inflammatory diseases. Inflamm Res 2025; 74:47. [PMID: 40047902 DOI: 10.1007/s00011-025-02021-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 02/11/2025] [Accepted: 03/02/2025] [Indexed: 05/13/2025] Open
Abstract
Nuclear factor-kappa B (NF-κB) transcription factor plays a crucial function in controlling several cellular processes, including the production of inflammatory mediators. The aberrant activation of this transcription factor and its signaling pathway is associated with the pathophysiology of many diseases. Therefore, discovering drugs that target NF-κB is crucial for treating various diseases. Decoy oligonucleotides (decoy ONs) are a pharmacological approach that specifically inhibits NF-κB activation and are used to treat several inflammatory diseases. Decoys that target NF-κB have been shown to enhance radiosensitivity and drug sensitivity in vitro and strongly block IL-6 and IL-8 gene expression induced by TNF-α in experimental cell systems. In vivo, NF-κB decoy reduced atherosclerotic plaque, prevented atopic dermatitis and extended cardiac transplant survival. Decoys have the potential to be used in clinical applications, but they face several challenges. To overcome these limitations, researchers have conducted studies on chemical modifications and delivery techniques. Innovative compounds that target NF-κB, such as NF-κB-decoy-based sensor-containing models, phosphorothioate hairpin-modified oligonucleotides, and peptide nucleic acid (PNA)-based transcription factor decoys, are very attractive. This research aims to explore the use of decoys to combat NF-κB in various disorders.
Collapse
Affiliation(s)
| | - Samaneh Rezaei
- Department of Medical Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alexandra E Butler
- Research Department, Royal College of Surgeons in Ireland Bahrain, Adliya, 15503, Bahrain
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Institute of Medical and Technical Sciences, Saveetha Medical College and Hospitals, Saveetha University, Chennai, India.
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
27
|
Zhang J, Shen M, Yin Y, Chen Y, Deng X, Mo J, Zhou X, Lin J, Chen X, Xie X, Wu X, Chen X. Carnosic acid reduces lipid content, enhances gut health, and modulates microbiota composition and metabolism in diet-induced obese mice. Food Funct 2025; 16:1888-1902. [PMID: 39932492 DOI: 10.1039/d4fo04534c] [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: 03/04/2025]
Abstract
Carnosic acid (CA) is a bioactive phenolic diterperne compound found in sage and rosemary. The present study investigated the beneficial effects of CA (50 and 100 mg per kg bw) in diet-induced obese mice and the underlying mechanisms of action. After the intervention, the physiology, lipid metabolism, and tissue morphology, as well as the inflammation, gut microbiota, and metabolomics in the colon were measured. We found that CA improved the composition and metabolism of the gut microbiota in obese mice, with Akkermansia being the dominant bacterium negatively correlated with obesity and various fecal metabolites. Regarding the intestinal barrier function, CA promoted the expression of tight junction proteins and inhibited the TLR4/MyD88/NF-κB signaling pathway in obese mice to alleviate colonic inflammation. These results suggest that CA improved multiple aspects of gut health in diet-induced obesity in mice, providing a scientific basis for future clinical studies in humans.
Collapse
Affiliation(s)
- Jing Zhang
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Mengzhu Shen
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Yue Yin
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Yuru Chen
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Xianying Deng
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Jingyun Mo
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Xiaoling Zhou
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Juanying Lin
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Xinxin Chen
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Xinwei Xie
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| | - Xian Wu
- Department of Kinesiology, Nutrition, and Health, Miami University, Oxford, Ohio 45056, USA.
| | - Xuexiang Chen
- School of Public Health, Guangzhou Medical University, Guangzhou 510642, Guangdong, P. R. China.
| |
Collapse
|
28
|
Forouzanfar F, Sahranavard T, Tsatsakis A, Iranshahi M, Rezaee R. Rutin: a pain-relieving flavonoid. Inflammopharmacology 2025; 33:1289-1301. [PMID: 39961908 DOI: 10.1007/s10787-025-01671-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 01/24/2025] [Indexed: 03/19/2025]
Abstract
Rutin (vitamin P or rutoside) is a citrus flavonoid glycoside that has shown beneficial health effects in different organs against various conditions including inflammation and pain. The majority of rutin therapeutic benefits are ascribed to its antioxidant and anti-inflammatory properties. This review article discusses studies that investigated pain-relieving activity of rutin and summarizes the reported mechanisms of action. Rutin pain-relieving effect has been studied in streptozotocin-induced diabetes, chronic constriction injury, and oxaliplatin-, formalin-, acetic acid- and glutamate-induced nociception in mice or rats. Based on the literature, rutin analgesic effects are induced through potentiation of antioxidant arsenal, reduction of inflammatory cytokines (e.g., Tumor necrosis factor alpha (TNF-α) and interleukin-1β) levels, suppression of cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS) expressions and modulation of MAPK, NF-κB and Nrf-2/HO-1 signaling. Preclinical findings on rutin pain-relieving activity are promising, however, its safety profile needs to be more thoroughly investigated and clinical trials should be conducted to assess its analgesic effects in humans.
Collapse
Affiliation(s)
- Fatemeh Forouzanfar
- Medical Toxicology Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Toktam Sahranavard
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Aristidis Tsatsakis
- Center of Toxicology Science & Research, Division of Morphology, Medical School, University of Crete, Voutes Campus, 71003, Heraklion, Greece
| | - Mehrdad Iranshahi
- Biotechnology Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Ramin Rezaee
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Center of Toxicology Science & Research, Division of Morphology, Medical School, University of Crete, Voutes Campus, 71003, Heraklion, Greece.
| |
Collapse
|
29
|
Xu J, Li N, Xie H, Duan C, Liao X, Li R, Zhang H, Pan Y, Ma X, Du S, Sheng J, Wang X, Yang L, Jin P. CSF3 promotes colorectal cancer progression by activating p65/NF-κB signaling pathway and inducing an immunosuppressive microenvironment. Transl Oncol 2025; 53:102310. [PMID: 39929064 PMCID: PMC11849657 DOI: 10.1016/j.tranon.2025.102310] [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: 03/07/2024] [Revised: 12/26/2024] [Accepted: 01/30/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Colony-stimulating factor 3 (CSF3) is a cytokine that promotes inflammation by stimulating the maturation, proliferation, and trafficking of myeloid progenitor cells. However, the functional importance of CSF3 in colorectal cancer (CRC) remains unclear. METHODS CSF3 expression levels in CRC cells and tissues were detected by quantitative real-time PCR (qRT-PCR), western blot and immunohistochemistry (IHC). In vitro and in vivo assays were performed to investigate the oncogenic function of CSF3 in the tumor associated malignant phenotypes and the tumorigenic capability of CRC cells. Immunocoprecipitation was performed to verify the regulatory effects of CSF3 on IκBα ubiquitination. RESULTS We found that CSF3 was overexpressed in CRC tissues compared to adjacent normal tissues, which correlated with poor patient survival. In vitro, silencing CSF3 significantly impaired cell proliferation, colony formation, and migration, while enhancing apoptosis. In vivo, silencing CSF3 resulted in reduced tumor growth, weight, and volume, indicating its potential as a therapeutic target. Mechanistically, CSF3 was found to mediate CRC development by activating the NF-κB signaling pathway, as evidenced by the decreased phosphorylation of p65 and reduced IκBα ubiquitination in CSF3-silenced cells. Furthermore, CSF3 silencing modulated immune infiltration in CRC, promoting an anti-tumor immune response and altering the tumor microenvironment. CONCLUSION CSF3 modulated the NF-κB signaling pathway through a distinct mechanism involving p65 phosphorylation and the activation of NF-κB by enhancing IκBα ubiquitination, thereby effectively promoting CRC development, and CSF3 may serve as a potential therapeutic target for repressing CRC advance and metastasis.
Collapse
Affiliation(s)
- Junfeng Xu
- Senior Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, PR China
| | - Na Li
- Senior Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, PR China; Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, 100700, PR China
| | - Hui Xie
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, 100700, PR China
| | - Changwei Duan
- Medical School of Chinese PLA, Beijing 100853, PR China
| | - Xingchen Liao
- Medical School of Chinese PLA, Beijing 100853, PR China
| | - Ruoran Li
- Medical School of Chinese PLA, Beijing 100853, PR China
| | - Heng Zhang
- Medical School of Chinese PLA, Beijing 100853, PR China
| | - Yuanming Pan
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing 101149, PR China
| | - Xianzong Ma
- Senior Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, PR China; Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, 100700, PR China
| | - Shuwen Du
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, 100700, PR China
| | - Jianqiu Sheng
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, 100700, PR China
| | - Xin Wang
- Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, 100700, PR China.
| | - Lang Yang
- Senior Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, PR China; Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, 100700, PR China.
| | - Peng Jin
- Senior Department of Gastroenterology, The First Medical Center of Chinese PLA General Hospital, Beijing 100853, PR China; Department of Gastroenterology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, 100700, PR China.
| |
Collapse
|
30
|
Son SK, Moon JS, Yang DW, Jung NR, Kang JH, Lee BN, Kim SH, Kim MS. Role of FOXO3a in LPS-induced inflammatory conditions in human dental pulp cells. J Oral Biosci 2025; 67:100614. [PMID: 39824385 DOI: 10.1016/j.job.2025.100614] [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: 09/13/2024] [Revised: 01/12/2025] [Accepted: 01/14/2025] [Indexed: 01/20/2025]
Abstract
OBJECTIVES We investigated the involvement of FOXO3a in lipopolysaccharide (LPS)-induced inflammation in primary human dental pulp cells (HDPCs). METHODS HDPCs that were isolated from donors undergoing tooth extraction for orthodontic purposes were cultured with or without 1 μg/mL LPS at various intervals. The FOXO3a localization in the HDPCs was verified using immunofluorescence. Proinflammatory cytokines, such as interleukin (IL) 1β, IL6, and IL8, as well as their underlying mechanisms were assessed by observing gene and protein expressions through quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot analyses. RESULTS LPS treatment enhanced the expressions of IL1β, IL6, and IL8 in HDPCs, concurrently activating nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB). Furthermore, FOXO3a expression was higher in the LPS-stimulated HDPCs, as confirmed by immunofluorescence localization. The results of loss-/gain-of-function approaches confirmed the regulatory role of FOXO3a in inflammatory HDPCs. FOXO3a knockdown attenuated proinflammatory cytokine expression; FOXO3a overexpression augmented their expression levels. FOXO3a inhibited retinoic acid receptor-related orphan receptor alpha (RORα) expression, thereby inactivating NFκB. CONCLUSION Our findings suggest that FOXO3a contributes to homeostasis in HDPCs through modulating the expression of proinflammatory cytokines under inflammatory conditions.
Collapse
Affiliation(s)
- Su-Kyung Son
- Dental Science Research Institute, School of Dentistry, Chonnam National University, Gwangju, South Korea
| | - Jung-Sun Moon
- Dental Science Research Institute, School of Dentistry, Chonnam National University, Gwangju, South Korea
| | - Dong-Wook Yang
- Dental Science Research Institute, School of Dentistry, Chonnam National University, Gwangju, South Korea
| | - Na-Ri Jung
- Dental Science Research Institute, School of Dentistry, Chonnam National University, Gwangju, South Korea
| | - Jee-Hae Kang
- Dental Science Research Institute, School of Dentistry, Chonnam National University, Gwangju, South Korea
| | - Bin-Na Lee
- Dental Science Research Institute, School of Dentistry, Chonnam National University, Gwangju, South Korea
| | - Sun-Hun Kim
- Dental Science Research Institute, School of Dentistry, Chonnam National University, Gwangju, South Korea
| | - Min-Seok Kim
- Dental Science Research Institute, School of Dentistry, Chonnam National University, Gwangju, South Korea.
| |
Collapse
|
31
|
Reichen C, Beirão BCB, Monteiro ALG. Contagious ecthyma in small ruminants: from etiology to vaccine challenges - a review. Vet Res Commun 2025; 49:115. [PMID: 39992468 DOI: 10.1007/s11259-025-10677-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 02/11/2025] [Indexed: 02/25/2025]
Abstract
Orf virus (ORFV) is an epitheliotropic, double-stranded DNA pathogen belonging to the genus Parapoxvirus, and it is the causative agent of contagious ecthyma (CE) in small ruminants. It is an endemic disease on goat and sheep herds around the world. It is often a neglected disease, with impacts on herd health and productivity, while also being an occupational zoonosis. This review explores the causative agent of ovine ecthyma, its epidemiology, and clinical manifestations, with a particular emphasis on its interaction with the host's immune system and the development of ORFV vaccines. Like other members of the Poxviridae family, ORFV expresses numerous immunomodulatory genes, which complicate vaccination efforts and disease management. This review highlights the challenges posed by ORFV in achieving effective immunization and discusses potential vaccine strategies to overcome these obstacles.
Collapse
Affiliation(s)
- Caroline Reichen
- Department of Animal Science, Sheep and Goat Production and Research Center (LAPOC), Federal University of Paraná (UFPR), Curitiba, Paraná, Brazil.
| | - Breno Castello Branco Beirão
- Department of Basic Pathology, Comparative Immunology Laboratory (LIC), Federal University of Paraná (UFPR), Curitiba, 81531-980, Paraná, Brazil
| | - Alda Lúcia Gomes Monteiro
- Department of Animal Science, Sheep and Goat Production and Research Center (LAPOC), Federal University of Paraná (UFPR), Curitiba, Paraná, Brazil
| |
Collapse
|
32
|
Mukaddam K, Ruggiero S, Berger SM, Cholewa D, Dekany G, Bartenstein A, Milošević M, Kühl S, Bornstein MM, Alhawasli F, Fasler-Kan E. TNF-α Activates NF-κB Signalling Pathway in MG-63 Cells on Titanium and Zirconia Implant Surfaces. MATERIALS (BASEL, SWITZERLAND) 2025; 18:884. [PMID: 40004407 PMCID: PMC11857784 DOI: 10.3390/ma18040884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/15/2025] [Accepted: 01/22/2025] [Indexed: 02/27/2025]
Abstract
Dental implant therapy is a widely used clinical procedure for restoring missing teeth in patients. Zirconia implants were introduced as an alternative to titanium implants due to their excellent biocompatibility and esthetic properties. The nuclear factor kappa B (NF-κB) signalling pathway is responsible for multiple aspects of innate and adaptive immune functions and serves as a significant and crucial mediator of inflammatory processes. The dysregulation of NF-κB activation induces pathological processes in multiple diseases. The purpose of this study was to investigate the activation of the NF-κB pathway upon stimulation with tumour necrosis factor (TNF)-α in osteoblast-like cells (MG-63) cultured on zirconia surfaces in comparison to titanium surfaces. Several methods such as immunoblot, immunofluorescence, MTT assay, and flow cytometry were used in this study. We observed that human recombinant TNF-α caused a strong activation of the NF-κB pathway on both zirconia and titanium discs and in wells without any discs. This activation was marked by the upregulation of MHC class I proteins in MG-63 cells grown on both titanium and zirconia discs; however, there was no effect on MHC class II protein expression. In summary, the present study has shown that TNF-α stimulation equally activates the NF-κB pathway in MG-63 cells cultured on both titanium and zirconia surfaces.
Collapse
Affiliation(s)
- Khaled Mukaddam
- Department of Oral Surgery, University Center for Dental Medicine Basel (UZB), University of Basel, Mattenstrasse 40, CH-4058 Basel, Switzerland; (K.M.)
| | - Sabrina Ruggiero
- Department of Pediatric Surgery, Children’s Hospital, Inselspital Bern, University of Bern, Freiburgstrasse 15, CH-3010 Bern, Switzerland
| | - Steffen M. Berger
- Department of Pediatric Surgery, Children’s Hospital, Inselspital Bern, University of Bern, Freiburgstrasse 15, CH-3010 Bern, Switzerland
| | - Dietmar Cholewa
- Department of Pediatric Surgery, Children’s Hospital, Inselspital Bern, University of Bern, Freiburgstrasse 15, CH-3010 Bern, Switzerland
| | - Gabriela Dekany
- Department of Pediatric Surgery, Children’s Hospital, Inselspital Bern, University of Bern, Freiburgstrasse 15, CH-3010 Bern, Switzerland
| | - Andreas Bartenstein
- Department of Pediatric Surgery, Children’s Hospital, Inselspital Bern, University of Bern, Freiburgstrasse 15, CH-3010 Bern, Switzerland
| | - Milan Milošević
- Department of Pediatric Surgery, Children’s Hospital, Inselspital Bern, University of Bern, Freiburgstrasse 15, CH-3010 Bern, Switzerland
| | - Sebastian Kühl
- Department of Oral Surgery, University Center for Dental Medicine Basel (UZB), University of Basel, Mattenstrasse 40, CH-4058 Basel, Switzerland; (K.M.)
| | - Michael M. Bornstein
- Department of Oral Health & Medicine, University Center for Dental Medicine Basel (UZB), University of Basel, Mattenstrasse 40, CH-4058 Basel, Switzerland
| | - Farah Alhawasli
- Department of Biomedicine, University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland
| | - Elizaveta Fasler-Kan
- Department of Pediatric Surgery, Children’s Hospital, Inselspital Bern, University of Bern, Freiburgstrasse 15, CH-3010 Bern, Switzerland
| |
Collapse
|
33
|
OKADA D, KANAI K, OTAKA Y, MATSUMOTO T, IZUMOTO A, UCHIYAMA Y, NAGAI N, YAMASHITA Y, ICHIKAWA Y, SUGIUCHI M, TAJIMA K. Anti-inflammatory effects of water-dispersible hesperetin on endotoxin-induced uveitis in rats involving the nuclear factor κB and Wnt/β-catenin signaling pathways. J Vet Med Sci 2025; 87:223-231. [PMID: 39814392 PMCID: PMC11830440 DOI: 10.1292/jvms.24-0453] [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: 11/06/2024] [Accepted: 12/25/2024] [Indexed: 01/18/2025] Open
Abstract
This study investigated the anti-inflammatory effects of water-dispersible hesperetin (WD-Hpt) in an endotoxin-induced uveitis (EIU) rat model. The rats were orally administered 10, 25, or 50 mg/kg WD-Hpt immediately after lipopolysaccharide (LPS) injection at the concentration of 200 μg. Clinical scores, cellular inflammation, the aqueous humor (ApH) protein concentration, as well as the levels of tumor necrosis factor (TNF)-α, cyclooxygenase (COX)-2 and inducible NO synthase (iNOS) in AqH, and histopathological grades were assessed. Immunohistostaining and mRNA analyses measured expressions of TNF-α, COX-2, iNOS, activated nuclear factor (NF)-κB p65, I kappa B (IκB)-α degradation, phosphorylated (p)-IκB kinase (IKK) α/β, β-catenin, and glycogen synthase kinase (GSK)-3β. Compared to LPS treated group (LPS txg), WD-Hpt treatment groups (WD-Hpt txg) resulted in the following results: 1) clinical scores improved [LPS txg; 3.90 ± 0.20, WD-Hpt txg; 2.40 ± 0.37 (P<0.05)], 2) the number of inflammatory cells in AqH decreased [LPS txg; 8.65 ± 1.41 × 105 cells/mL, WD-Hpt txg; 3.83 ± 1.20 × 105 cells/mL (P<0.05)], 3) AqH protein concentration reduced [LPS txg; 36.65 ± 2.71 mg/mL, WD-Hpt txg; 28.73 ± 2.36 mg/mL (P<0.05)], and 4) decreased levels of TNF-α [LPS txg; 69.55 ± 7.38 pg/mL, WD-Hpt txg; 35.18 ± 9.22 pg/mL (P<0.001)], iNOS [LPS txg; 153.37 ± 12.72 μM, WD-Hpt txg; 110.79 ± 13.27 μM (P<0.05)], and COX-2 [LPS txg; 1,080.56 ± 196.06 pg/mL, WD-Hpt txg; 477.80 ± 66.61 pg/mL (P<0.01)] in AqH were observed, and histopathological grades improved [LPS txg; 2.80 ± 0.40, WD-Hpt txg; 1.50 ± 0.50 (P<0.05)]. Immunostaining and mRNA analysis revealed that 50 mg/kg WD-Hpt effectively suppressed iNOS, COX-2, NF-κB p65, IκB-α degradation, p-IKKα/β, β-catenin, and GSK-3β expression. These findings suggested that WD-Hpt exerts anti-inflammatory effects by targeting the NF-κB and Wnt/β-catenin pathways.
Collapse
Affiliation(s)
- Daiki OKADA
- Department of Small Animal Internal Medicine II, School of
Veterinary Medicine, Kitasato University, Aomori, Japan
| | - Kazutaka KANAI
- Department of Small Animal Internal Medicine II, School of
Veterinary Medicine, Kitasato University, Aomori, Japan
| | - Yuya OTAKA
- Department of Small Animal Internal Medicine II, School of
Veterinary Medicine, Kitasato University, Aomori, Japan
| | - Tsubasa MATSUMOTO
- Department of Small Animal Internal Medicine II, School of
Veterinary Medicine, Kitasato University, Aomori, Japan
| | - Akane IZUMOTO
- Department of Small Animal Internal Medicine II, School of
Veterinary Medicine, Kitasato University, Aomori, Japan
| | - Yumiko UCHIYAMA
- Department of Small Animal Internal Medicine II, School of
Veterinary Medicine, Kitasato University, Aomori, Japan
| | - Noriaki NAGAI
- Faculty of Pharmacy, Kindai University, Osaka, Japan
| | - Yohei YAMASHITA
- Department of Small Animal Internal Medicine II, School of
Veterinary Medicine, Kitasato University, Aomori, Japan
| | - Yoichiro ICHIKAWA
- Department of Small Animal Internal Medicine II, School of
Veterinary Medicine, Kitasato University, Aomori, Japan
| | - Misaki SUGIUCHI
- Department of Small Animal Internal Medicine II, School of
Veterinary Medicine, Kitasato University, Aomori, Japan
| | - Kazuki TAJIMA
- Department of Small Animal Internal Medicine II, School of
Veterinary Medicine, Kitasato University, Aomori, Japan
| |
Collapse
|
34
|
Sun Y, Sen S, Parmar R, Arakawa-Hoyt J, Cappelletti M, Rossetti M, Gjertson DW, Sigdel TK, Sarwal MM, Schaenman JM, Bunnapradist S, Lanier LL, Pickering H, Reed EF. Cytotoxic KLRG1+ IL-7R- effector CD8+ T cells distinguish kidney transplant recipients controlling cytomegalovirus reactivation. Front Immunol 2025; 16:1542531. [PMID: 40028342 PMCID: PMC11868092 DOI: 10.3389/fimmu.2025.1542531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 01/27/2025] [Indexed: 03/05/2025] Open
Abstract
Introduction Cytomegalovirus (CMV) viremia remains a major contributor to clinical complications in solid organ transplant (SOT) patients, including organ injury, morbidity and mortality. Given their critical role in antiviral defense, CD8+ T cells are essential for protective immunity against CMV. Methods Using single-cell RNA sequencing, we investigated the transcriptional signatures and developmental lineages of CD8+ T cells in eight immunosuppressed kidney transplant recipients (KTRs) who received organs from CMV-seropositive donors. Results were validated in a cohort of 62 KTRs using immunophenotyping. Results Our data revealed a significant influence of CMV serostatus on transcriptional variance of CD8+ memory T cells, associating with the first principal component from a global analysis of CD8+ T cells (p =0.0406), forming a continuum with five principal differentiation trajectories driven by CMV primary infection or reactivation. Following CMV primary infection, CD8+ T cells were hallmarked by restrained effector-memory differentiation. CD8+ T cells during CMV reactivation diverged non-linearly into senescent-like cells with signatures of arrested cell cycle, diminished translational activity and downregulated ZNF683 and longitudinally expanding effector cells with robust cytotoxic potential and upregulated ZNF683, acting as a reservoir for long-lived effector cells supporting long-term protection. Notably, CD28lo KLRG1hi IL-7R (CD127)lo HLA-DRhi CD8+ T cells present prior to the detection of viremia in CMV-seropositive patients emerged as a key feature distinguishing patients who did or did not undergo CMV reactivation after prophylaxis discontinuation (p =0.0163). Frequencies of these cells were also positively correlated with CMV-stimulated secretion of IFN-γ (p =0.0494), TNF-α (p =0.0358), MIP-1α (p =0.0262), MIP-1β (p =0.0043). Discussion These results provide insights into the transcriptional regulation that influences the generation of CD8+ T cell immunity to CMV and may inform strategics for monitoring host immune response to CMV to better identify and introduce therapeutic intervention to patients at risk of developing clinically significant CMV viremia.
Collapse
Affiliation(s)
- Yumeng Sun
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Subha Sen
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Rajesh Parmar
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Janice Arakawa-Hoyt
- Department of Microbiology and Immunology, Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, CA, United States
| | - Monica Cappelletti
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Maura Rossetti
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - David W. Gjertson
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Tara K. Sigdel
- Department of Surgery, Division of Multi Organ Transplantation, University of California, San Francisco, San Francisco, CA, United States
| | - Minnie M. Sarwal
- Department of Surgery, Division of Multi Organ Transplantation, University of California, San Francisco, San Francisco, CA, United States
| | - Joanna M. Schaenman
- Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Suphamai Bunnapradist
- Division of Nephrology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Lewis L. Lanier
- Department of Microbiology and Immunology, Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, CA, United States
| | - Harry Pickering
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Elaine F. Reed
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
35
|
Zhang Y, Xie J, Feng Y, Qadeer A, Li S, Deng X, Zhu L, Kong B, Xia Z. Post-translational modifications as a key mechanism for herpes simplex virus type I evasion of host innate immunity. Front Microbiol 2025; 16:1543676. [PMID: 40008039 PMCID: PMC11850380 DOI: 10.3389/fmicb.2025.1543676] [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: 12/11/2024] [Accepted: 01/24/2025] [Indexed: 02/27/2025] Open
Abstract
Herpes simplex virus type 1 (HSV-1) is a DNA virus that infects humans and establishes long-term latency within the host. Throughout its prolonged interaction with the host, HSV-1 evades the innate immune system by encoding its own proteins. Post-translational modifications (PTMs) of these proteins play crucial roles in their function, activity, and interactions with other factors by modifying specific amino acids, thereby enabling a diverse range of protein functions. This review explores the mechanisms and roles of PTMs in HSV-1-encoded proteins, such as phosphorylation, ubiquitination, deamidation, and SUMOylation, during HSV-1 infection and latency. These modifications are essential for suppressing host innate immunity, facilitating viral replication, and elucidating the crosstalk among various post-translational modifications.
Collapse
Affiliation(s)
- Yongxing Zhang
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
| | - Junlei Xie
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
| | - Ying Feng
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
| | - Abdul Qadeer
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
| | - Shanni Li
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
| | - Xu Deng
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Lipeng Zhu
- School of Life Sciences, Xiangya School of Medicine, Central South University, Changsha, China
| | - Bo Kong
- China Tobacco Hunan Industrial, Changsha, China
| | - Zanxian Xia
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
- Hunan Key Laboratory of Animal Models for Human Diseases, Hunan Key Laboratory of Medical Genetics and Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, China
| |
Collapse
|
36
|
Ma X, Mei S, He Y, Wuyun Q, Zhou L, Cai Z, Luo Q, Wen Y, Yan J. Unraveling the association and regulatory role of miR-146b-5p in coronary artery disease. BMC Cardiovasc Disord 2025; 25:81. [PMID: 39910430 PMCID: PMC11796014 DOI: 10.1186/s12872-025-04530-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 01/28/2025] [Indexed: 02/07/2025] Open
Abstract
BACKGROUND Coronary artery disease (CAD), one of the most prevalent cardiovascular diseases, is a critical health issue that affects millions of individuals worldwide. It has been reported that miR-146b-5p exhibited a strong correlation with inflammatory responses and atherosclerosis. However, its association with the incidence and severity of CAD has not been substantiated in a large cohort. In the study, we focus on the expression of miR-146b-5p in peripheral blood mononuclear cells (PBMCs) of patients with CAD and preliminarily investigate its function and underlying mechanism. METHODS AND RESULTS The study encompassed a total of 452 participants, consisting 295 patients with CAD and 157 individuals without CAD. Quantitative reverse transcription-polymerase chain reaction (qRT-PCR) was performed to assess miR-146b-5p expression in PBMCs. We found that miR-146b-5p was significantly increased in PBMCs of patients with CAD compared with the control group. Binary logistic regression revealed that miR-146b-5p was associated with CAD. Receiver Operation Characteristic (ROC) analysis showed that the sensitivity and specificity of miR-146b-5p in discriminating CAD patients from non-CAD patients were meaningful. Subsequent subgroup analysis showed that miR-146b-5p was related to the severity of CAD. Furthermore, gain- and loss-of-function experiments in THP-1 cells showed that miR-146b-5p inhibited inflammation, cell proliferation, and migration. Mechanically, miR-146b-5p was involved in the classical NF-κB inflammatory pathway by directly targeting IKKβ. CONCLUSION Our study revealed that miR-146b-5p was higher in the PBMCs of CAD patients than non-CAD individuals, and established a correlation between miR-146b-5p and occurrence and severity of CAD. In addition, the inflammatory role of miR-146b-5p is mediated by targeting IKKβ.
Collapse
Affiliation(s)
- Xiaozhu Ma
- Department of Cardiology, Division of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Shuai Mei
- Department of Cardiology, Division of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Yi He
- Department of Cardiology, Division of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Qidamugai Wuyun
- Department of Cardiology, Division of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Li Zhou
- Department of Cardiology, Division of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Ziyang Cai
- Department of Cardiology, Division of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Qiushi Luo
- Department of Cardiology, Division of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Yi Wen
- Department of Cardiology, Division of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Jiangtao Yan
- Department of Cardiology, Division of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China.
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China.
| |
Collapse
|
37
|
Kang X, Han Y, Wu M, Li Y, Qian P, Xu C, Zou Z, Dong J, Wei J. In situ blockade of TNF-TNFR2 axis via oncolytic adenovirus improves antitumor efficacy in solid tumors. Mol Ther 2025; 33:670-687. [PMID: 39690741 PMCID: PMC11853363 DOI: 10.1016/j.ymthe.2024.12.011] [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: 02/21/2024] [Revised: 07/13/2024] [Accepted: 12/10/2024] [Indexed: 12/19/2024] Open
Abstract
Tumor necrosis factor (TNF) has been recognized as an immune activation factor in tumor immunotherapy. Our study demonstrated that TNF blockade markedly enhanced the antitumor efficacy of oncolytic adenovirus (AdV) therapy. To minimize systemic side effects, we engineered a recombinant oncolytic AdV encoding a TNF inhibitor (AdV-TNFi) to confine TNF blockade within the tumor microenvironment (TME). AdV-TNFi significantly improved therapeutic outcomes across various solid tumor models, including four murine and two golden hamster cancers. Immune cell profiling identified CD8+ T cells as the primary mediators of AdV-TNFi-induced antitumor effects, rather than CD4+ T or NK cells. Additionally, AdV-TNFi significantly decreased the infiltration of suppressive myeloid-derived immune cells within the TME and promoted long-term antitumor immune surveillance. Further investigation indicated that TNFR2, more than TNFR1, is pertinent to the immunosuppressive TME, with a recombinant AdV-encoding anti-TNFR2 demonstrating comparable antitumor efficacy to AdV-TNFi. Moreover, AdV-TNFi enhanced the antitumor efficacy of gemcitabine and immune checkpoint blockades (ICBs), such as anti-PD-L1 and anti-TIGIT antibodies, in pancreatic carcinoma and the anti-EGFR antibody in colon carcinoma. In conclusion, intratumoral blockade of the TNF/TNFR2 axis using AdV augments cancer immunotherapy efficacy while mitigating the risks associated with systemic TNF or TNFR2 suppression, warranting further clinical investigation.
Collapse
MESH Headings
- Humans
- Animals
- Cricetinae
- Cell Line
- Male
- Mice
- Mice, Inbred C57BL
- Adenoviridae/genetics
- Tumor Necrosis Factors/chemistry
- Tumor Necrosis Factors/genetics
- Tumor Necrosis Factors/metabolism
- Receptors, Tumor Necrosis Factor, Type II/chemistry
- Receptors, Tumor Necrosis Factor, Type II/genetics
- Receptors, Tumor Necrosis Factor, Type II/metabolism
- Neoplasms/genetics
- Neoplasms/metabolism
- Neoplasms/pathology
- Disease Progression
- Mice, Inbred BALB C
- Receptors, Tumor Necrosis Factor, Type I/genetics
- Receptors, Tumor Necrosis Factor, Type I/metabolism
- Lymphocytes, Tumor-Infiltrating/immunology
- CD8-Positive T-Lymphocytes/immunology
- Protein Binding
- Models, Molecular
- Protein Structure, Tertiary
- Protein Structure, Quaternary
- Immune Checkpoint Inhibitors/pharmacology
- Tyrosine Kinase Inhibitors/pharmacology
Collapse
Affiliation(s)
- Xiaozhen Kang
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Yifeng Han
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Mengdi Wu
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Yuxin Li
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Peng Qian
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Chuning Xu
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Zhengyun Zou
- The Comprehensive Cancer Center of Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jie Dong
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, China; Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, Jiangsu 226001, China.
| | - Jiwu Wei
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, China.
| |
Collapse
|
38
|
Langhnoja J, Buch L, Pillai P. Neurotrophomodulatory effect of TNF-α through NF-κB in rat cortical astrocytes. Cytotechnology 2025; 77:37. [PMID: 39776978 PMCID: PMC11700960 DOI: 10.1007/s10616-024-00698-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: 04/21/2024] [Accepted: 12/24/2024] [Indexed: 01/11/2025] Open
Abstract
Tumor necrosis factor alpha (TNF-α) is a well-known pro-inflammatory cytokine originally recognized for its ability to induce apoptosis and cell death. However, recent research has revealed that TNF-α also plays a crucial role as a mediator of cell survival, influencing a wide range of cellular functions. The signaling of TNF-α is mediated through two distinct receptors, TNFR1 and TNFR2, which trigger various intracellular pathways, including NF-κB, JNK, and caspase signaling cascades. Both TNFR1 and TNFR2 are expressed in astrocytes, which are specialized glial cells essential for maintaining the structural and functional integrity of the central nervous system (CNS). Astrocytes support neuronal function by regulating brain homeostasis, maintaining synaptic function, and supplying metabolic substrates. In addition, astrocytes are known to secrete a variety of growth factors and neurotrophins, such as nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), neurotrophin-3 (NT-3), and NT-4/5. These neurotrophins play a critical role in supporting neuronal survival, synaptic plasticity, and myelination within the brain. The present study focuses on the role of TNF-α in modulating neurotrophin expression and secretion in rat cortical astrocytes. We demonstrate that TNF-α induces the upregulation of neurotrophins, particularly NGF and BDNF, in cultured astrocytes. This effect is accompanied by an increase in the expression of their respective receptors (TrkA & TrkB), further suggesting a functional modulation of neurotrophic signaling pathways. Notably, we show that the modulation of neurotrophin expression by TNF-α is mediated via the NF-κB signaling pathway. Additionally, we observed that TNF-α also regulates the secretion levels of NGF and BDNF into the culture media of astrocytes in a dose-dependent manner, indicating that TNF-α can modulate both the production and release of these growth factors. Taken together, our findings highlight a previously underexplored neuroprotective role of TNF-α in astrocytes. Specifically, we propose that TNF-α, through the upregulation of neurotrophins, may contribute to maintaining neuronal health and supporting neuroprotection under disease conditions.
Collapse
Affiliation(s)
- Jaldeep Langhnoja
- Division of Neurobiology, Department of Zoology, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat India
| | - Lipi Buch
- Division of Neurobiology, Department of Zoology, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat India
| | - Prakash Pillai
- Division of Neurobiology, Department of Zoology, The Maharaja Sayajirao University of Baroda, Vadodara, Gujarat India
| |
Collapse
|
39
|
Ma C, Jiang Y, Xiang Y, Li C, Xie X, Zhang Y, You Y, Xie L, Gong J, Sun Y, Tong S, Song Q, Chen J, Xiao W. Metabolic Reprogramming of Macrophages by Biomimetic Melatonin-Loaded Liposomes Effectively Attenuates Acute Gouty Arthritis in a Mouse Model. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410107. [PMID: 39717013 PMCID: PMC11831490 DOI: 10.1002/advs.202410107] [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: 08/22/2024] [Revised: 11/25/2024] [Indexed: 12/25/2024]
Abstract
Gouty arthritis is characterized by an acute inflammatory response triggered by monosodium urate (MSU) crystals deposited in the joints and periarticular tissues. Current treatments bring little effects owing to serious side effects, necessitating the exploration of new and safer therapeutic options. Macrophages play a critical role in the initiation, progression, and resolution of acute gout, with the cellular profiles closely linked to their activation and polarization. This suggests that metabolic regulation can be of significance in managing gouty inflammation. In this study, it is demonstrated that melatonin, a natural hormone, modulates the metabolic remodeling of inflammatory macrophages by shifting their metabolism from glycolysis to oxidative phosphorylation, further altering functions of the pathogenic macrophage. To improve melatonin delivery to the inflamed sites, macrophage membrane-coated melatonin-loaded liposomes (MLT-MLP) are developed. Benefiting from the inflammation-homing characteristic of macrophage membrane, such engineered liposomes effectively target the inflamed site and demonstrate potent anti-inflammatory effects, achieving an enhanced amelioration of acute gouty arthritis. In conclusion, this study proposes a novel strategy aimed at metabolic reprogramming of macrophages to attenuate the pathological injuries in acute gout, providing a potential therapeutic strategy of gout-associated diseases, especially gouty arthritis.
Collapse
Affiliation(s)
- Chuchu Ma
- Department of Pharmaceutics, School of Pharmacy & Shanghai Pudong Hospital, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, China
| | - Yuyu Jiang
- Department of Pathogen Biology, Naval Medical University, Shanghai, 200433, China
| | - Yan Xiang
- Department of Pathogen Biology, Naval Medical University, Shanghai, 200433, China
| | - Chang Li
- Department of Pharmaceutics, School of Pharmacy & Shanghai Pudong Hospital, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, China
| | - Xiaoying Xie
- Department of Pharmaceutics, School of Pharmacy & Shanghai Pudong Hospital, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, China
| | - Yunkai Zhang
- Naval Medical Center, Naval Medical University, Shanghai, 200433, China
| | - Yang You
- Department of Pharmaceutics, School of Pharmacy & Shanghai Pudong Hospital, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, China
| | - Laozhi Xie
- Department of Pharmaceutics, School of Pharmacy & Shanghai Pudong Hospital, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, China
| | - Jianing Gong
- Department of Pharmaceutics, School of Pharmacy & Shanghai Pudong Hospital, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, China
| | - Yinzhe Sun
- Department of Pharmaceutics, School of Pharmacy & Shanghai Pudong Hospital, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, China
| | - Shiqiang Tong
- Department of Pharmaceutics, School of Pharmacy & Shanghai Pudong Hospital, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, China
| | - Qingxiang Song
- Department of Pharmacology and Chemical Biology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Universities Collaborative Innovation Center for Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jun Chen
- Department of Pharmaceutics, School of Pharmacy & Shanghai Pudong Hospital, Key Laboratory of Smart Drug Delivery, Ministry of Education, Fudan University, Shanghai, 201203, China
| | - Wenze Xiao
- Department of Rheumatology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| |
Collapse
|
40
|
Shen G, Wang G, Chen J, Guo Y, Zhang W, Xu C, Chen L, Wang Q. MALT1 promotes the antibacterial immune response by activating NF-κB signaling and enhancing hemocyte phagocytosis in the Chinese mitten crab. FISH & SHELLFISH IMMUNOLOGY 2025; 157:110100. [PMID: 39733914 DOI: 10.1016/j.fsi.2024.110100] [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: 11/11/2024] [Revised: 12/19/2024] [Accepted: 12/25/2024] [Indexed: 12/31/2024]
Abstract
Mucosa-associated lymphoid tissue lymphoma translocation protein 1 (MALT1), a scaffold protein, plays a pivotal role in the NF-κB pathway downstream of T-cell receptors (TCRs) and B-cell receptors (BCRs). As a key signaling hub, MALT1 integrates various pathways, making it essential for both innate and adaptive immunity. However, its role in the antibacterial immune responses of crustaceans remains unclear. Here, we characterized MALT1 from the Chinese mitten crab (Eriocheir sinensis), denoted as EsMALT1, and compared its sequence and domain conservation with MALT1 from other species. Furthermore, Vibrio parahaemolyticus infection upregulated EsMALT1 expression markedly. Knockdown of EsMALT1 in hemocytes inhibits the translocation of the NF-κB-like transcription factors EsRelish and EsDorsal from the cytoplasm to the nucleus in response to Vibrio parahaemolyticus stimulation, thereby reducing the expression of the antimicrobial peptides anti-lipopolysaccharide factor (ALF), and Crustins. At the cellular level, silencing of EsMALT1 expression significantly inhibited the phagocytic capacity of crab hemocytes against Vibrio parahaemolyticus. In vivo, silencing of EsMALT1 rendered crabs susceptible to bacterial infection and impaired their bacterial clearance. In conclusion, EsMALT1 promotes both humoral and cellular immunity in E. sinensis, making it essential for the induction of antibacterial immune responses.
Collapse
Affiliation(s)
- Guoqing Shen
- School of Life Sciences, East China Normal University, Shanghai, China
| | - Guangyu Wang
- School of Aquatic and Life Sciences, Shanghai Ocean University, Shanghai, China
| | - Jinming Chen
- School of Aquatic and Life Sciences, Shanghai Ocean University, Shanghai, China
| | - Yanan Guo
- School of Life Sciences, East China Normal University, Shanghai, China
| | - Wen Zhang
- School of Life Sciences, East China Normal University, Shanghai, China
| | - Chaohui Xu
- School of Aquatic and Life Sciences, Shanghai Ocean University, Shanghai, China
| | - Liqiao Chen
- School of Life Sciences, East China Normal University, Shanghai, China.
| | - Qun Wang
- School of Aquatic and Life Sciences, Shanghai Ocean University, Shanghai, China.
| |
Collapse
|
41
|
Zhu X, Jia Y, Zhao Z, Zhang X, Zhao Y, Gui S, Yang XA. Cell signaling communication within papillary craniopharyngioma revealed by an integrated analysis of single-cell RNA sequencing and bulk RNA sequencing. J Transl Med 2025; 23:124. [PMID: 39871369 PMCID: PMC11773883 DOI: 10.1186/s12967-025-06149-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 01/18/2025] [Indexed: 01/29/2025] Open
Abstract
OBJECTIVE This study aims to elucidate the primary signaling communication among papillary craniopharyngioma (PCP) tumor cells. METHODS Five samples of PCP were utilized for single-cell RNA sequencing. The most relevant ligand and receptor interactions among different cells were calculated using the CellChat package in R software. Bulk RNA sequencing of 11 tumor samples and five normal controls was used to investigate the pair interactions detected by single-cell RNA sequencing. RESULTS Fibroblasts were not found in ACP, whereas they were detected in PCP. InferCNV revealed high CNV scores for the clusters of epithelial cells and fibroblasts using immune cells as a reference. Epithelial Mesenchymal Transition, Interferon Gamma Response, p53 Pathway, and Estrogen Response Early are pathways commonly shared by fibroblasts and epithelial cells, ranking high in priority. The Wnt signaling pathway and PI3K-Akt signaling pathway play a crucial role in facilitating communication between epithelial cells and fibroblasts. Neutrophils were recognized as the main receivers of incoming signals, with ANXA1-FPR1 and MIF-(CD74 + CXCR2) being identified as the primary signals transmitted from fibroblasts to neutrophils. CONCLUSION Through analyzing the communication of essential signaling pathways, ligands, and receptors among epithelial cells, fibroblasts, and neutrophils in PCP tumor tissues, we have identified certain molecules with promising prognostic and therapeutic potential.
Collapse
Affiliation(s)
- Xiaoyue Zhu
- Laboratory of Gene Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, Chengde, 067000, China
- Graduate School of Chengde Medical University, Chengde, 067000, China
- Department of Biomedical Engineering, Chengde Medical University, Chengde, 067000, China
| | - Yanfei Jia
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zicheng Zhao
- Laboratory of Gene Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, Chengde, 067000, China
| | - Xiaoyu Zhang
- Laboratory of Gene Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, Chengde, 067000, China
| | - Yunlong Zhao
- Laboratory of Gene Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, Chengde, 067000, China
| | - Songbai Gui
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Xiu-An Yang
- Laboratory of Gene Engineering and Genomics, School of Basic Medical Sciences, Chengde Medical University, Chengde, 067000, China.
- Hebei Key Laboratory of Nerve Injury and Repair, Chengde Medical University, Chengde, China.
| |
Collapse
|
42
|
Li YZ, Wang C, Peng X, Wang B, Wang JS, Xie HT, Zhang MC. Caffeine's protective role in dry eye disease and meibomian gland dysfunction: insights from clinical and experimental models. Int Immunopharmacol 2025; 146:113863. [PMID: 39709912 DOI: 10.1016/j.intimp.2024.113863] [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: 11/04/2024] [Revised: 11/30/2024] [Accepted: 12/11/2024] [Indexed: 12/24/2024]
Abstract
PURPOSE Inflammation and apoptosis contribute to the development of dry eye disease (DED) and meibomian gland dysfunction (MGD). This study aimed to investigate the effect of caffeine on the ocular surface and tear inflammatory cytokines through clinical, in vivo, and in vitro experiments. METHODS In the clinical study, comprehensive ophthalmic examinations of participants in the control and the caffeine groups were compared, including ocular surface and tears inflammatory cytokines. For in vitro study, rat meibomian gland epithelial cells (RMGECs) and human corneal epithelial cells (HCECs) were pretreated with or without caffeine and then stimulated with lipopolysaccharide (LPS). Inflammatory responses, apoptosis, and differentiation in cells were analyzed. In vivo study, apolipoprotein E knockout (ApoE-/-) mice were given caffeine-diet or no caffeine-diet, and their meibomian glands (MGs) and corneal tissue were compared. RESULTS Participants in the caffeine group exhibited significantly healthier ocular surface, lower tears inflammatory cytokines and a reduced prevalence of DED compared to the control group. In vitro study, caffeine pretreatment attenuated inflammatory responses, apoptosis and differentiation in LPS-induced RMGECs. Meanwhile, caffeine also markedly suppressed inflammatory responses and apoptosis in LPS-induced HCECs. In vivo study showed that ApoE-/- mice with caffeine-diet had more normal morphology of MGs and corneas compared to those without caffeine-diet, along with reduced inflammatory responses, cells apoptosis and ductal keratinization. Both in vitro and in vivo studies indicated that caffeine treatment was observed to inactivate of nuclear factor kappa B (NF-κB) phosphorylation. CONCLUSIONS Our study indicated that caffeine may be a protective potential of ocular surface, providing a new perspective on clinical treatment for DED and MGD.
Collapse
Affiliation(s)
- Yu-Zhi Li
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chao Wang
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xi Peng
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bei Wang
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia-Song Wang
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hua-Tao Xie
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Ming-Chang Zhang
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
43
|
Zhendong Y, Changjun C, Haocheng H, Qibin L, Dailing C, Linsong T, Xuecheng S, Gong M, Lei Z. Regulation of macrophage polarization and pyroptosis by 4-methylcatechol alleviates collagen-induced arthritis via Nrf2/HO-1 and NF-κB/NLRP3 signaling pathways. Int Immunopharmacol 2025; 146:113855. [PMID: 39709906 DOI: 10.1016/j.intimp.2024.113855] [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: 10/18/2024] [Revised: 12/09/2024] [Accepted: 12/11/2024] [Indexed: 12/24/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disease that leads to joint deformity and ultimately disability. The metabolite of quercetin, 4-Methylcatechol (4-MC), has been acknowledged for its anti-inflammatory and antioxidant properties; however, the protective effects of 4-MC on RA and its underlying mechanisms remain incompletely elucidated. In a collagen-induced arthritis (CIA) model, we observed that 4-MC effectively mitigated joint inflammation and bone destruction in CIA mice. Additionally, it significantly suppressed the upregulated expression of inflammatory cytokines in synovial tissues. Mechanistically, upon lipopolysaccharide (LPS) stimulation, 4-MC inhibited M1 polarization of macrophages and induced a phenotypic switch from M1 to M2 phenotype, thereby reducing the release of pro-inflammatory cytokines by M1 macrophages while increasing the release of anti-inflammatory cytokines by M2 macrophages. Furthermore, it attenuated LPS/adenosine triphosphate (ATP)-induced pyroptosis in macrophages by downregulating NLRP3 expression levels along with cleaved caspase-1, cleaved IL-1β, and GSDMD-NT expression levels. Notably, our findings revealed that 4-MC exerted inhibitory effects on the NF-κB signaling pathway through specific modulation of the NF-κB complex as well as phosphorylation of the upstream IKK kinase complex. Collectively, these results highlight significant therapeutic potential for utilizing 4-MC in RA treatment.
Collapse
Affiliation(s)
- Ying Zhendong
- Department of Orthopaedics Surgery, Shandong Provincial Qianfoshan Hospital, Shandong University of Traditional Chinese Medicine, Jinan 250012, PR China.
| | - Chen Changjun
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250012, PR China.
| | - Hou Haocheng
- The First Clinical College, Shandong University, Jinan 250014, PR China.
| | - Liu Qibin
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250012, PR China.
| | - Chen Dailing
- Department of Orthopaedics Surgery, Shandong Provincial Qianfoshan Hospital, Shandong Second Medical University, Jinan 250012, PR China.
| | - Teng Linsong
- Department of Orthopaedics Surgery, Shandong Provincial Qianfoshan Hospital, Shandong University of Traditional Chinese Medicine, Jinan 250012, PR China.
| | - Sun Xuecheng
- Department of Orthopedic Trauma, Weifang People's Hospital, Weifang, Shandong, PR China.
| | - Mouchun Gong
- Department of General Surgery, The First People's Hospital of Hangzhou Lin'an District (Hangzhou Medical College Affiliated Lin'an People's Hospital), Hangzhou 311300, PR China.
| | - Zhang Lei
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan 250012, PR China.
| |
Collapse
|
44
|
Eigenbrood J, Wong N, Mallory P, Pereira J, Morris-II DW, Beck JA, Cronk JC, Sayers CM, Mendez M, Kaiser L, Galindo J, Singh J, Cardamone A, Pore M, Kelly M, LeBlanc AK, Cotter J, Kaplan RN, McEachron TA. Spatial profiling identifies regionally distinct microenvironments and targetable immunosuppressive mechanisms in pediatric osteosarcoma pulmonary metastases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.22.631350. [PMID: 39896512 PMCID: PMC11785069 DOI: 10.1101/2025.01.22.631350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Osteosarcoma is the most common malignant bone tumor in young patients and remains a significant clinical challenge, particularly in the context of metastatic disease. Despite extensive documentation of genomic alterations in osteosarcoma, studies detailing the immunosuppressive mechanisms within the metastatic osteosarcoma microenvironment are lacking. Our objective was to characterize the spatial transcriptional landscape of metastatic osteosarcoma to reveal these immunosuppressive mechanisms and identify promising therapeutic targets. Here, we performed spatial transcriptional profiling on a cohort of osteosarcoma pulmonary metastases from pediatric patients. We reveal a conserved spatial gene expression pattern resembling a foreign body granuloma, characterized by peripheral inflammatory signaling, fibrocollagenous encapsulation, lymphocyte exclusion, and peritumoral macrophage accumulation. We also show that the intratumoral microenvironment of these lesions lack inflammatory signaling. Additionally, we identified CXCR4 as an actionable immunomodulatory target that bridges both the intratumoral and extratumoral microenvironments and highlights the spatial heterogeneity and complexity of this pathway. Collectively, this study reveals that metastatic osteosarcoma specimens are comprised of multiple regionally distinct immunosuppressive microenvironments.
Collapse
Affiliation(s)
- Jason Eigenbrood
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
- Current Address: University of Cambridge, Cancer Research UK Cambridge Institute, Cambridge, UK
- These authors contributed equally to this manuscript
| | - Nathan Wong
- Collaborative Bioinformatics Resource, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
- These authors contributed equally to this manuscript
| | - Paul Mallory
- Imaging Mass Cytometry Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Janice Pereira
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Douglass W Morris-II
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Jessica A Beck
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - James C Cronk
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Carly M Sayers
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Monica Mendez
- Department of Pathology and Laboratory Medicine, Children’s Hospital Los Angeles, Los Angeles, CA, USA
| | - Linus Kaiser
- Department of Pathology and Laboratory Medicine, Children’s Hospital Los Angeles, Los Angeles, CA, USA
| | - Julie Galindo
- Department of Pathology and Laboratory Medicine, Children’s Hospital Los Angeles, Los Angeles, CA, USA
| | - Jatinder Singh
- Center for Cancer Research Single Cell Analysis Facility, Cancer Research Technology Program, Frederick National Laboratory, Bethesda, MD, USA
| | - Ashley Cardamone
- Imaging Mass Cytometry Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Milind Pore
- Imaging Mass Cytometry Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Michael Kelly
- Center for Cancer Research Single Cell Analysis Facility, Cancer Research Technology Program, Frederick National Laboratory, Bethesda, MD, USA
| | - Amy K LeBlanc
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Jennifer Cotter
- Department of Pathology and Laboratory Medicine, Children’s Hospital Los Angeles, Los Angeles, CA, USA
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Rosandra N Kaplan
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Troy A McEachron
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
45
|
Zeng Y, Wu Z, Xiong M, Liang Z, Chen Z, Huang H, Yang H, Chen Q. Piezo1 promotes vibration-induced vascular smooth muscle injury by regulating the NF-κB/p65 axis. Commun Biol 2025; 8:96. [PMID: 39833492 PMCID: PMC11747106 DOI: 10.1038/s42003-025-07524-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 01/13/2025] [Indexed: 01/22/2025] Open
Abstract
Vibration induced damage to the peripheral circulatory system is thought to be an early stage of hand-arm vibration syndrome (HAVS) caused by occupational exposure to hand-transmitted vibration (HTV). This study investigated the mechanisms underlying vibration-induced vascular injury, focusing on the role of Piezo1, a mechanosensitive channel, and its association with the NF-κB/p65 signaling pathway. We demonstrated that vibration exposure leads to Piezo1-mediated upregulation of angiogenic chemokines, including CCL2, CCL5, CXCL1, CXCL2, and CXCL10, through the NF-κB/p65 pathway. To mimic the effects of vibration, a rat vibration model and a cellular vibration model were used. Animal and cellular models showed that vibration-induced vascular dysfunction while increasing Piezo1 expression. Piezo1 knockdown or p65 inhibition attenuated these effects, suggesting a crucial role for the Piezo1-NF-κB/p65 axis in vascular dysfunction. Furthermore, chemokines were identified as potential biomarkers for early diagnosis of HAVS in occupationally exposed individuals. These results highlight Piezo1 and the NF-κB/p65 pathway as potential therapeutic targets for HAVS and underscore the need for further validation in human samples and exploration of additional signaling mechanisms involved in vibration-induced vascular injury.
Collapse
Affiliation(s)
- Yingshan Zeng
- Department of Occupational and Environmental Health, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment (2019GCZX012), Guangdong Pharmaceutical University, Guangdong, China
| | - Zhiquan Wu
- Department of Occupational and Environmental Health, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment (2019GCZX012), Guangdong Pharmaceutical University, Guangdong, China
| | - Mengtian Xiong
- Department of Occupational and Environmental Health, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment (2019GCZX012), Guangdong Pharmaceutical University, Guangdong, China
| | - Zhishan Liang
- Department of Occupational and Environmental Health, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment (2019GCZX012), Guangdong Pharmaceutical University, Guangdong, China
| | - Ziyu Chen
- Department of Occupational and Environmental Health, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment (2019GCZX012), Guangdong Pharmaceutical University, Guangdong, China
| | - Huimin Huang
- Department of Occupational and Environmental Health, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment (2019GCZX012), Guangdong Pharmaceutical University, Guangdong, China
| | - Hongyu Yang
- Department of Occupational and Environmental Health, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China.
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment (2019GCZX012), Guangdong Pharmaceutical University, Guangdong, China.
| | - Qingsong Chen
- Department of Occupational and Environmental Health, School of Public Health, Guangdong Pharmaceutical University, Guangzhou, China.
- Guangdong Provincial Engineering Research Center of Public Health Detection and Assessment (2019GCZX012), Guangdong Pharmaceutical University, Guangdong, China.
| |
Collapse
|
46
|
He Y, Liu Y, Zhang M. The beneficial effects of curcumin on aging and age-related diseases: from oxidative stress to antioxidant mechanisms, brain health and apoptosis. Front Aging Neurosci 2025; 17:1533963. [PMID: 39906716 PMCID: PMC11788355 DOI: 10.3389/fnagi.2025.1533963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 01/06/2025] [Indexed: 02/06/2025] Open
Abstract
Aging and age-related disease are among the most common and challenging issues worldwide. During the aging process, the accumulation of oxidative stress, DNA damage, telomere dysfunction, and other related changes lead to cellular dysfunction and the development of diseases such as neurodegenerative and cardiovascular conditions. Curcumin is a widely-used dietary supplement against various diseases such as cancer, diabetes, cardiovascular diseases and aging. This agent mediates its effects through several mechanisms, including the reduction of reactive oxygen species (ROS) and oxidative stress-induced damage, as well as the modulation of subcellular signaling pathways such as AMPK, AKT/mTOR, and NF-κB. These pathways are involved in cellular senescence and inflammation, and their modulation can improve cell function and help prevent disease. In cancer, Curcumin can induce apoptosis in a variety of different tumor cell lines. Curcumin also activates redox reactions within cells inducing ROS production that leads to the upregulation of apoptosis receptors on the tumor cell membrane. Curcumin can also upregulate the expression and activity of p53 that inhibits tumor cell proliferation and increases apoptosis. Furthermore, curcumin has a potent inhibitory effect on the activity of nuclear factor kappa B (NF-κB) and cyclooxygenase-2 (COX-2), which are involved in the overexpression of antiapoptosis genes such as Bcl-2. It can also attenuate the regulation of antiapoptosis phosphoinositide 3-kinases (PI3K) signaling and increase the expression of mitogen-activated protein kinases (MAPKs) to induce endogenous production of ROS. Therefore, herein, we aim to summarize how curcumin affect different epigenetic processes (such as apoptosis and oxidative stress) in order to change aging-related mechanisms. Furthermore, we discuss its roles in age-related diseases, such as Alzheimer, Parkinson, osteoporosis, and cardiovascular diseases.
Collapse
Affiliation(s)
- Ying He
- Department of Biological and Food Engineering, Lyuliang University, Lishi, Shanxi, China
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi, China
| | - Yongqing Liu
- Department of Biological and Food Engineering, Lyuliang University, Lishi, Shanxi, China
| | - Min Zhang
- Key Laboratory of Agro-Products Primary Processing, Academy of Agricultural Planning and Engineering, MARA, Beijing, China
| |
Collapse
|
47
|
Yuan J, Liu C, Jiang C, Liu N, Yang Z, Xing H. RSL3 induces ferroptosis by activating the NF-κB signalling pathway to enhance the chemosensitivity of triple-negative breast cancer cells to paclitaxel. Sci Rep 2025; 15:1654. [PMID: 39794456 PMCID: PMC11724089 DOI: 10.1038/s41598-025-85774-w] [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: 06/03/2024] [Accepted: 01/06/2025] [Indexed: 01/13/2025] Open
Abstract
Chemotherapy resistance in triple-negative breast cancer (TNBC) leads to poor therapeutic effects and a poor prognosis. Given that paclitaxel-based chemotherapy is the main treatment method for TNBC, enhancing its chemosensitivity has been a research focus. Induced ferroptosis of tumour cells has been proven to increase chemosensitivity, but its ability to sensitize TNBC cells to paclitaxel (PTX) is unknown. In our experiments, measurements of viability and proliferation validated the synergistic effect of PTX combined with RSL3 on TNBC cells. The accumulation of intracellular Fe2+ and lipid reactive oxygen species, as well as the expression of malondialdehyde, illustrated that RSL3 enhanced the chemosensitivity of TNBC to PTX by inducing ferroptosis. Through transcriptome sequencing, a series of differentially expressed genes were identified, in which the expression of cytokines, such as CXCLs, was significantly increased in the treatment group, and the effect of combination therapy on TNBC was enriched mainly in the NFκB signalling pathway. In subsequent validation experiments, the use of the NF-κB inhibitor BAY11-7082 reversed the inhibitory effects of PTX and RSL3 on TNBC cell activity. In a xenograft immunodeficient mouse model, the inhibitory effects of PTX and RSL3 on TNBC in vivo were further verified. Our research validated the synergistic effects of PTX and RSL3 both in vivo and in vitro, with RSL3 inducing ferroptosis by activating the NF-κB signalling pathway, thereby increasing the chemosensitivity of TNBC to PTX. This study provides new insights for improving the therapeutic efficacy of treatment strategies.
Collapse
Affiliation(s)
- Jialin Yuan
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Cong Liu
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Chengwei Jiang
- Department of Pathology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Ning Liu
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun, China.
| | - Zhaoying Yang
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun, China.
| | - Hua Xing
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun, China.
| |
Collapse
|
48
|
Miao L, Cheong MS, Zhang H, Khan H, Tao H, Wang Y, Cheang WS. Portulaca oleracea L. (purslane) extract ameliorates intestinal inflammation in diet-induced obese mice by inhibiting the TLR4/NF-κB signaling pathway. Front Pharmacol 2025; 15:1474989. [PMID: 39845784 PMCID: PMC11752911 DOI: 10.3389/fphar.2024.1474989] [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: 08/02/2024] [Accepted: 12/04/2024] [Indexed: 01/24/2025] Open
Abstract
Background Portulaca oleracea L. (purslane) is a dietary plant and a botanical drug with antioxidant, antidiabetic, and anti-inflammatory activities. However, the effects of purslane against intestinal-inflammation-associated obesity are yet to be studied. In the present study, we hypothesized that purslane extract could reduce intestinal inflammation associated with metabolic disorder. Results Male C57BL/6J mice were fed a high-fat diet (HFD, 60% kcal% of fat) for a total duration of 14 weeks to establish an obesity model; further, the treatment group was orally administered purslane extract (200 mg/kg/day) during the last 4 weeks. Then, intestinal tissues were detached from the mice for detecting protein expressions through Western blot and immunohistochemical analyses. Pro-inflammatory cytokines were determined using ELISA kits, whereas the components of purslane extract were detected by ultra performance liquid chromatography/electrospray ionization quadrupole time-of-flight mass spectrometry. Chronic oral administration of purslane extract ameliorated colon shortening syndrome and reduced bowel inflammation in HFD-induced obese mice through suppression of the toll-like receptor 4 (TLR4)/nuclear factor kappa B (NF-κB) signaling pathway to downregulate TLR4, myeloid differentiation factor 88 (MyD88), Ser32 phosphorylation of NF-κB inhibitor alpha (IκBα), and Ser536 phosphorylation of NF-κB p65 expression levels, thereby inhibiting the pro-inflammatory cytokines, tumor necrosis factor (TNF)-α and interleukin (IL)-6 levels. Conclusion The present study supports the anti-inflammatory potential of purslane extract for modulating bowel inflammation under obesity through inhibition of the TLR4/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Lingchao Miao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | - Meng Sam Cheong
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | - Haolin Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan, Pakistan
- Department of Pharmacy, Korea University, Sejong, South Korea
| | - Hongxun Tao
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
| | - Yuxiao Wang
- College of Food Science and Engineering, Shandong Agricultural University, Tai’an, Shandong, China
| | - Wai San Cheang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| |
Collapse
|
49
|
Dhariwal R, Dave K, Jain M. Omics-based analysis of mitochondrial dysfunction and BBB integrity in post-COVID-19 sequelae. Sci Rep 2024; 14:31016. [PMID: 39730725 DOI: 10.1038/s41598-024-82180-6] [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: 12/03/2024] [Indexed: 12/29/2024] Open
Abstract
The SARS-CoV-2 virus that resulted in the COVID-19 pandemic has been implicated in a range of neurological issues, such as encephalopathy, stroke, and cognitive decline. Although the precise mechanism causing these issues is unknown, mounting evidence shows that blood-brain barrier (BBB) disruption is probable2 a major factor. The integrity of the blood-brain barrier (BBB), a highly selective barrier that divides the brain from the systemic circulation, is crucial for preserving normal brain function. By analysing the multi-transcriptome data, this work explores the neurological impacts of the SARS-CoV-2 virus and provides insight into the molecular mechanisms behind BBB breakdown and neurological symptoms in COVID-19 patients. The endothelial cells of BBB expresses inflammatory genes in response to the systemic inflammation induced due to SARS-CoV-2 remnants in the body. This raises the possibility that systemic inflammation brought on by SARS-CoV-2 and BBB integrity are correlated. Furthermore, the study highlights the pathways involved in oxidative stress and endothelial cell activation, revealing their role in COVID-19 passage through BBB and induction of systemic inflammation and advancement toward neurological disorders. The article showcases the evidence that mitochondrial dysfunction is a major aftermath associated with SARS-CoV-2 infection as the impaired Mitochondria leads to an accumulation of reactive oxygen species (ROS), triggering endothelial dysfunction, and leading to the passage of harmful molecules across the BBB. This study offers insightful information that may open up the possibilities for new treatment plans by targeting biomarkers specifically associated with inflammation and BBB dysfunctioning conditions.
Collapse
Affiliation(s)
- Rupal Dhariwal
- Cell and Developmental Biology Laboratory, Research and Development Cell, PIMSR, Parul University, Vadodara, Gujarat, 391760, India
- Parul Institute of Applied Sciences, Department of Life Sciences, Parul University, Vadodara, 391760, Gujarat, India
| | - Kirtan Dave
- Bioinformatics Laboratory, Research & Development Cell, Parul University, Vadodara, Gujarat, 391760, India.
- Parul Institute of Paramedical and Health Sciences Faculty of Medicine, Parul University,, Vadodara, Gujarat-391760, India.
| | - Mukul Jain
- Cell and Developmental Biology Laboratory, Research and Development Cell, PIMSR, Parul University, Vadodara, Gujarat, 391760, India.
- Parul Institute of Applied Sciences, Department of Life Sciences, Parul University, Vadodara, 391760, Gujarat, India.
| |
Collapse
|
50
|
Han H, Gao Y, Chen B, Xu H, Shi C, Wang X, Liang Y, Wu Z, Wang Z, Bai Y, Wu C. Nrf2 inhibits M1 macrophage polarization to ameliorate renal ischemia-reperfusion injury through antagonizing NF-κB signaling. Int Immunopharmacol 2024; 143:113310. [PMID: 39383788 DOI: 10.1016/j.intimp.2024.113310] [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: 05/02/2024] [Revised: 09/13/2024] [Accepted: 09/30/2024] [Indexed: 10/11/2024]
Abstract
Renal ischemia-reperfusion injury (IRI) is a condition that arises from a sudden interruption of the blood flow to the kidney for a period of time followed by restoration of the blood supply. This process contributes to acute kidney injury (AKI), increases morbidity and mortality, and is a major risk factor for chronic kidney disease (CKD). Nuclear factor erythroid-derived 2-like 2 (Nrf2) has been shown to exhibit strong anti-oxidative and anti-inflammatory effects, which are reciprocally regulated by the pro-inflammatory actions of nuclear factor-kappa B (NF-κB) signaling. In this study, we established a model of AKI caused by renal IRI in mice lacking the Nrf2 gene (KO-Nrf2) and mice pre-injected with ML385 (Nrf2 inhibitor). In addition, LPS- or IL-4-induced M1- or M2-type polarized macrophages (RAW264.7), respectively, were also treated with Nrf2 activation and inhibition. The results demonstrated a more pronounced activation of the NF-κB signaling pathway in the Nrf2 inhibition model, accompanied by a more severe inflammatory effect. In cultured macrophages and renal IRI mice, Nrf2 inhibition activated M1 macrophage polarization, thereby increasing the release of proinflammatory cell factors (iNOS and TNF-α) and aggravating renal IRI. Notably, the inhibitory effect of Nrf2 on M1 macrophage polarization was related to the downregulation of the NF-κB signaling pathway activity, resulting in partial relief of renal IRI. Consequently, our findings indicated that Nrf2 inhibits M1 macrophage polarization to ameliorate renal IRI through antagonizing NF-κB signaling. Targeted activation of Nrf2 may be one of the important strategies for renal IRI treatment.
Collapse
Affiliation(s)
- Hui Han
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Yuanyuan Gao
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Boxuan Chen
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Hongjie Xu
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Chenghao Shi
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Xiaowu Wang
- Department of Burns and Skin Repair Surgery, The Third Affiliated Hospital, Wenzhou Medical University, Ruian 325200, Zhejiang Province, China
| | - Yihan Liang
- Department of Urology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai 317000, China
| | - Zhixuan Wu
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Ziqiong Wang
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Yongheng Bai
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China.
| | - Cunzao Wu
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China.
| |
Collapse
|