Mengfei Ma, Jianxun Guo, Xiaoying Su, Baocai Ma, Xiufen Wang, Mingyu Wangshao, Kai Zhong, Yueying Wang, Guoyu Yang, Yingqian Han
{"title":"Aryl hydrocarbon receptor (AhR) alleviates the LPS-induced inflammatory responses in IPEC-J2 cells by activating PINK1/Parkin-mediated mitophagy.","authors":"Mengfei Ma, Jianxun Guo, Xiaoying Su, Baocai Ma, Xiufen Wang, Mingyu Wangshao, Kai Zhong, Yueying Wang, Guoyu Yang, Yingqian Han","doi":"10.1007/s00011-025-02063-y","DOIUrl":"https://doi.org/10.1007/s00011-025-02063-y","url":null,"abstract":"<p><strong>Objective: </strong>This study investigates the role of the aryl hydrocarbon receptor (AhR) in lipopolysaccharide (LPS)-induced inflammatory responses in IPEC-J2 cells.</p><p><strong>Methods: </strong>Inflammatory responses were triggered in IPEC-J2 cells using 5 μg/ml LPS. AhR was activated with tryptophan or FICZ and knocked down via RNA interference. PINK1/Parkin-mediated mitophagy was activated using CCCP and inhibited by PINK1 knockdown. Inflammatory mediators and pathway proteins were analyzed through ELISA, RT-qPCR, western blot, and immunofluorescence. Mitochondrial function was assessed by measuring ROS, ATP, and mitochondrial membrane potential. The interaction between AhR and PINK1 was examined using dual-luciferase reporter assays.</p><p><strong>Results: </strong>The IDO1/AhR signaling axis was activated in LPS-stimulated IPEC-J2 cells. AhR activation was found to attenuate LPS-induced inflammatory responses, whereas AhR knockdown exacerbated these responses. Mechanistic investigations demonstrated that AhR activation alleviated LPS-induced mitochondrial damage. Activating PINK1/Parkin-mediated mitophagy successfully countered the increased inflammatory response in IPEC-J2 cells after AhR knockdown. Moreover, blocking PINK1 reversed the anti-inflammatory effects of FICZ. Dual-luciferase reporter assays revealed that AhR acts as a crucial transcription factor by directly binding to the promoter region, thereby initiating PINK1 transcription.</p><p><strong>Conclusions: </strong>AhR reduces LPS-induced inflammatory response in IPEC-J2 cells by activating PINK1/Parkin-mediated mitophagy, with AhR directly engaging the PINK1 promoter to enhance its transcription. Targeting AhR may present a novel strategy for the prevention and management of Escherichia coli-induced diarrhea in piglets.</p>","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":"74 1","pages":"98"},"PeriodicalIF":4.8,"publicationDate":"2025-06-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144527774","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Clinical outcomes of anti-inflammatory therapies inhibiting the NLRP3/IL-1β/IL-6/CRP pathway in coronary artery disease patients: a systemic review and meta-analysis of 37,056 individuals from 32 randomized trials.","authors":"Yannan Pan, Fangfang Fan, Jie Jiang, Yan Zhang","doi":"10.1007/s00011-025-02058-9","DOIUrl":"10.1007/s00011-025-02058-9","url":null,"abstract":"<p><strong>Background: </strong>Treatment effects of anti-inflammatory therapies inhibiting the NLRP3/IL-1β/IL-6/CRP pathway in coronary artery disease (CAD) had conflicting results. The study aims to evaluate efficacy and safety outcomes of treatments inhibiting this pathway.</p><p><strong>Methods: </strong>Cochrane Library, Embase, Pubmed, and ClinicalTrials.gov were searched for randomized controlled trials evaluating therapies inhibiting the NLRP3/IL-1β/IL-6/CRP pathway in CAD patients. Relative risks (RR) with 95% confidence intervals (CI) were calculated.</p><p><strong>Results: </strong>32 studies and 37,056 individuals were included. Anti-inflammatory therapies inhibiting the pathway reduced the risks of myocardial infarction (MI) (RR 0.85, 95% CI 0.78-0.93) and coronary revascularization (RR 0.80, 95% CI 0.74-0.86), with no benefits in major adverse cardiovascular events (MACE), heart failure (HF), stroke, cardiovascular or all-cause mortality. Colchicine reduced the risks of MACE, MI, and coronary revascularization. IL-1 inhibitors reduced the risks of coronary revascularization, with potential benefits in MI and HF. Increased risks of infections, gastrointestinal adverse effects, and injection site reactions were found. Meta-regression analysis demonstrated that post-treatment hsCRP/CRP was correlated with MACE (p < 0.001) and MI (p = 0.048) and post-treatment IL-6 was associated with MI (p = 0.033).</p><p><strong>Conclusion: </strong>Anti-inflammatory therapies inhibiting the NLRP3/IL-1β/IL-6/CRP pathway had satisfying safety profiles and were beneficial in preventing MI and coronary revascularization in CAD patients despite no benefits in stroke, cardiovascular, or all-cause mortality.</p>","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":"74 1","pages":"99"},"PeriodicalIF":4.8,"publicationDate":"2025-06-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12206679/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144527776","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Zhonghao Li, Shengsong Chen, Nan Gao, Jie Chen, Ying Qin, Guoqiang Zhang
{"title":"Identification of key genes and development of an identifying machine learning model for sepsis.","authors":"Zhonghao Li, Shengsong Chen, Nan Gao, Jie Chen, Ying Qin, Guoqiang Zhang","doi":"10.1007/s00011-025-02068-7","DOIUrl":"https://doi.org/10.1007/s00011-025-02068-7","url":null,"abstract":"<p><strong>Objective and design: </strong>This study aims to identify key genes of sepsis and construct a model for sepsis identification through integrated multi-organ single-cell RNA sequencing (scRNA-seq) and machine learning.</p><p><strong>Material or subjects: </strong>Datasets downloaded from the Gene Expression Omnibus (GSE207363, GSE207651, GSE185263, GSE69063 and GSE134347) were used.</p><p><strong>Methods: </strong>ScRNA-seq data extracted from heart (GSE207363) and lung tissues (GSE207651) of septic mice were processed and analyzed using the Seurat package in R. Key genes were identified as present in both heart and lung tissues, resulting from the overlap of three analyses along with differential expression analyses. We then used support vector machine recursive feature elimination to construct a model for sepsis identification based on these key genes. The GSE185263 dataset was used for training, while GSE69063 and GSE134347 were used for testing. The accuracy of the model in identifying of sepsis was validated by analyzing the area under the receiver operating characteristic curve (AUROC) using the test datasets.</p><p><strong>Results: </strong>Thirteen genes were initially identified as key genes, and after translation to their human homologs, ten genes remained. The optimal SVM-RFE model incorporated eight of these genes (CAMP, CD74, HLA-DQA1, HLA-DQB1, HLA-DMA, HLA-DRB5, and LYZ). In the two test datasets, the AUROC value for the accuracy of the model in identifying of sepsis was 0.904 and 0.924, respectively.</p><p><strong>Conclusions: </strong>We have identified several key genes and developed a machine learning model for sepsis identification. Further studies are needed to validate our findings.</p>","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":"74 1","pages":"100"},"PeriodicalIF":4.8,"publicationDate":"2025-06-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144527778","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Óscar Gorgojo-Galindo, María Álvarez-Bardón, Adrián García-Concejo, Rocío López-Herrero, María Heredia-Rodríguez, Eduardo Tamayo, Hugo Gonzalo-Benito
{"title":"Biological attributes of zinc-dependent endopeptidases in endothelial dysfunction associated with sepsis: a narrative review.","authors":"Óscar Gorgojo-Galindo, María Álvarez-Bardón, Adrián García-Concejo, Rocío López-Herrero, María Heredia-Rodríguez, Eduardo Tamayo, Hugo Gonzalo-Benito","doi":"10.1007/s00011-025-02056-x","DOIUrl":"https://doi.org/10.1007/s00011-025-02056-x","url":null,"abstract":"<p><p>Understanding of the pathophysiology of sepsis allows the development of new therapeutic approaches, beyond supportive care and antimicrobial therapy, to help reduce the high mortality rates in intensive care units. In this context, a large number of experimental and clinical studies in the last 20 years support the key role of the endothelium in sepsis in relation to organ failure. This review aims to provide a comprehensive overview of the role of zinc-dependent endopeptidases and their endogenous inhibitors in endothelial dysfunction associated with sepsis as well as in other pathologies. Finally, we discuss that inhibitors of this family of proteases could represent a promising therapeutic approach against sepsis.</p>","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":"74 1","pages":"95"},"PeriodicalIF":4.8,"publicationDate":"2025-06-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144527775","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"MitoQ alleviates mitochondria damage in sepsis-acute lung injury in a citrate synthase dependent manner.","authors":"Jiaojiao Sun, Sihao Jin, Zhiqiang Wang","doi":"10.1007/s00011-025-02055-y","DOIUrl":"10.1007/s00011-025-02055-y","url":null,"abstract":"<p><p>Sepsis is a systemic inflammatory disease caused by severe infection, involving multiple organs in the body, with the lungs being the most susceptible, leading patients to develop acute lung injury (ALI). Mitoquinone Mesylate (MitoQ) is an antioxidant specifically designed to target mitochondria, and it has anti-aging and antioxidant properties. This study aimed to investigate the protective effects of MitoQ on sepsis-induced ALI and its mechanisms. C57BL/6 mice were used to establish the cecal ligation and puncture (CLP) model of sepsis and were orally administered or not administered MitoQ for two weeks. MitoQ effectively alleviated sepsis-induced lung tissue damage, inflammatory responses, oxidative stress, and apoptosis. Furthermore, MitoQ significantly inhibited oxidative stress and mitochondrial damage in pulmonary macrophages. Mechanistically, MitoQ upregulated the mRNA and protein levels of citrate synthase (CS) in lung tissues and pulmonary macrophages. Silencing the CS gene with siRNA significantly reduced the protective effects of MitoQ against oxidative stress, inflammation, and cell apoptosis. In conclusion, MitoQ alleviates sepsis-induced ALI by preserving mitochondrial function.</p>","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":"74 1","pages":"92"},"PeriodicalIF":4.8,"publicationDate":"2025-06-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12187896/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144475062","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Exosomes as immunomodulators in autoimmune inflammation: implications for primary Sjögren's disease.","authors":"Yanggang Hong, Siyan Chen, Xiaoyang Jiang, Jinwen Zhang, Xinyue Liang, Jiani Yao, Sheng Gao, Chunyan Hua","doi":"10.1007/s00011-025-02053-0","DOIUrl":"https://doi.org/10.1007/s00011-025-02053-0","url":null,"abstract":"<p><p>Primary Sjögren's disease (pSD) is a systemic autoimmune disorder characterized by exocrine gland dysfunction and lymphocytic infiltration, leading to dry mouth and eyes. Increasing evidence implicates extracellular vesicles, particularly exosomes, as critical mediators of immune regulation in pSD. This review outlines the biogenesis, molecular composition, and immunomodulatory functions of exosomes, and summarizes their emerging roles in the pathogenesis, diagnosis, and potential treatment of pSD. Exosomes derived from immune and glandular cells carry diverse cargoes, such as miRNAs, proteins, and nucleic acids, that modulate disease-relevant immune pathways. For example, exosomal miR-BART13-3p from Epstein-Barr virus-infected B cells suppresses STIM1 and AQP5, impairing salivary gland function, while PD-L1-enriched exosomes from mesenchymal stem cells inhibit T follicular helper cell polarization via the PI3K/AKT pathway, thereby modulating B-cell activation. Additional exosomal cargoes affect Th17/Treg balance, inflammasome activation, and type I interferon signaling. We also highlight the diagnostic potential of disease-specific exosomal markers in plasma and discuss advances in preclinical studies using exosomes as therapeutic agents. However, significant challenges remain, including cargo heterogeneity, lack of standardized isolation methods, and limited clinical data, all of which hinder the translation of exosome-based therapies into clinical practice. By integrating mechanistic and translational findings, this review provides a comprehensive perspective on the immunological and clinical relevance of exosomes in pSD. These insights may guide future development of diagnostic biomarkers and targeted therapies in autoimmune diseases.</p>","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":"74 1","pages":"91"},"PeriodicalIF":4.8,"publicationDate":"2025-06-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144283819","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Abbas Shahi, Zahra Firoozi, Ghaidaa Raheem Lateef Al-Awsi, Ebrahim Mirzaei, Hojjat Shahbazi, Zahra Rezaee, Elham Mohammadisoleimani, Yaser Mansoori, Ali Moravej
{"title":"The relationship between neurodevelopmental disorders (NDDs) and NLRP3 inflammasome.","authors":"Abbas Shahi, Zahra Firoozi, Ghaidaa Raheem Lateef Al-Awsi, Ebrahim Mirzaei, Hojjat Shahbazi, Zahra Rezaee, Elham Mohammadisoleimani, Yaser Mansoori, Ali Moravej","doi":"10.1007/s00011-025-02052-1","DOIUrl":"https://doi.org/10.1007/s00011-025-02052-1","url":null,"abstract":"<p><p>The brain's process of creating neural networks that affect functionality or performance is referred to as neurodevelopment. A person's capacity to learn and remember, pay attention, regulate their emotions, socialize, exercise and self-control are just a few of the cognitive and motor abilities that can be affected by abnormal brain development. Numerous neurodevelopmental diseases have been functionally related to abnormalities in innate immune signaling networks. Innate immunity is in charge of the defense of humans against pathogens and tissue damage by triggering inflammation, mainly through sensing pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs). Some of these inflammatory processes have been shown to be mediated by large multiprotein complexes called inflammasomes which are present in cytosol and play important roles in the immune system. Due to its role in defense against infectious agents, bacteria, viruses, and fungi, NLRP3 is almost the most studied and well-known member of inflammasomes. The involvement of NLRP3 in inflammatory illnesses is evident due to the wide range of triggers and its intricate signaling pathways. In disorders with neurodevelopmental underpinnings, such as autism spectrum disorder (ASD), attention deficit hyperactivity disorder (ADHD), schizophrenia (SCZ), obsessive compulsive disorder (OCD), bipolar disorder (BD), Tourette syndrome (TS), etc., neuroinflammation plays a significant role in their pathophysiology. For these reasons, we reviewed the roles of NLRP3 in various neurodevelopmental disorders below.</p>","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":"74 1","pages":"90"},"PeriodicalIF":4.8,"publicationDate":"2025-06-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144208452","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Jiawen Zhang, Chengfeng Zhang, Zhongyi Xu, Jun Liang, Qinyuan Zhu
{"title":"Successful treatment of refractory erythrodermic pemphigus foliaceus with low-dose rituximab and intravenous immunoglobulin.","authors":"Jiawen Zhang, Chengfeng Zhang, Zhongyi Xu, Jun Liang, Qinyuan Zhu","doi":"10.1007/s00011-025-02059-8","DOIUrl":"https://doi.org/10.1007/s00011-025-02059-8","url":null,"abstract":"<p><strong>Background: </strong>Erythrodermic pemphigus foliaceus (EPF) is a severe, often refractory autoimmune disease.</p><p><strong>Method: </strong>We report successful treatment of refractory EPF in a 59-year-old male using low-dose rituximab (RTX) and intravenous immunoglobulin (IVIG).</p><p><strong>Results: </strong>Initial high-dose corticosteroids and IVIG failed, complicated by infections. A modified regimen of RTX (0.5 g) and IVIG (0.4 g/kg/day for three days) every two weeks for two cycles induced remission, with a mild relapse at six months successfully retreated. No infections occurred during 15-month follow-up.</p><p><strong>Conclusion: </strong>This combined therapy using low-dose RTX and IVIG may be a safe and effective option for refractory EPF, potentially minimizing infection risk associated with standard RTX dosing. Further studies are warranted.</p>","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":"74 1","pages":"89"},"PeriodicalIF":4.8,"publicationDate":"2025-06-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144208451","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
M Dhilsath Fathima, S P Raja, K Jayanthi, R Hariharan
{"title":"OptiStack classifier: optimized stacking framework with ensemble feature engineering for enhanced cardiovascular risk prediction.","authors":"M Dhilsath Fathima, S P Raja, K Jayanthi, R Hariharan","doi":"10.1007/s00011-025-02039-y","DOIUrl":"https://doi.org/10.1007/s00011-025-02039-y","url":null,"abstract":"<p><strong>Background: </strong>Cardiovascular diseases (CVD) are a leading cause of morbidity and mortality globally, highlighting the urgent need for accurate risk prediction to improve early intervention and management. Traditional models have difficulty capturing the complex interactions between risk factors, which limits their predictive power.</p><p><strong>Objective: </strong>This paper proposes the OptiStack Classifier, an optimized stacking framework developed to enhance CVD risk prediction through ensemble feature engineering and machine learning techniques.</p><p><strong>Methods: </strong>The model uses dimensionality reduction and ensemble feature engineering methods, including polynomial expansion, binning and domain-specific feature transformation, to improve data representation. Principal Component Analysis (PCA) is used to dimensionality reduction, improving computational efficiency. A stacking framework integrates multiple machine learning algorithms as base learners, with Logistic Regression acting as the meta-classifier. Bayesian Optimization is applied for hyperparameter tuning, further boosting predictive performance.</p><p><strong>Results: </strong>The proposed model shows significant improvements in predicting CVD risk, helping with early diagnosis and prevention, which can lead to better health outcomes for patients.</p>","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":"74 1","pages":"88"},"PeriodicalIF":4.8,"publicationDate":"2025-05-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144191732","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Jingwen Zhang, Muhammad Abid Hayat, Yu Si, Tao Guo, Yinying Ni, Qian Wang, Yancheng Hong, Yudie Cao, Sijia He, Zijuan Weng, Fengmei Li, Hao Zuo, Xin Sun, Bo Chen, Jiabo Hu
{"title":"Interleukin-37 modulates microglial phenotype and inhibits inflammatory response via the MyD88/NF-κB pathway in lipopolysaccharide-induced neuroinflammation.","authors":"Jingwen Zhang, Muhammad Abid Hayat, Yu Si, Tao Guo, Yinying Ni, Qian Wang, Yancheng Hong, Yudie Cao, Sijia He, Zijuan Weng, Fengmei Li, Hao Zuo, Xin Sun, Bo Chen, Jiabo Hu","doi":"10.1007/s00011-025-02048-x","DOIUrl":"https://doi.org/10.1007/s00011-025-02048-x","url":null,"abstract":"<p><strong>Objective: </strong>Interleukin-37 (IL-37), an anti-inflammatory cytokine within the interleukin-1 (IL-1) family, exhibits immunomodulatory properties. Here we evaluate the effects of IL-37 on microglia in neuroinflammation and its potential mechanisms.</p><p><strong>Methods: </strong>C57BL/6 mice were injected intraperitoneally with 1 µg of recombinant human IL-37 protein (rhIL-37), and 24 h later with lipopolysaccharide (LPS) (5 mg/kg) to induce neuroinflammation. After 2-h pretreatment of BV2 cells with rhIL-37 (100 ng/mL), an in vitro model was established by treating with LPS (100 ng/mL). Mice were assessed for behavioral tests, and neuronal damage was evaluated by Nissl staining and hematoxylin and eosin staining. The expression of Iba1, CD86, CD206, and NF-κB were detected by immunofluorescence staining, and inflammatory mediators and pathway proteins were evaluated by ELISA, qRT-PCR, and Western blot.</p><p><strong>Results: </strong>IL-37 significantly ameliorated LPS-induced behavioral deficits and protected mice from inflammatory injury. In vitro experiments suggested that IL-37 modulates polarization of microglia from M1 to M2 phenotype, along with reducing pro-inflammatory cytokine production. Moreover, IL-37 attenuated the production of NF-κB and MyD88.</p><p><strong>Conclusions: </strong>IL-37 regulates microglia against neuroinflammatory responses by blocking the MyD88/NF-κB pathway and shows for the first time how IL-37 influences the phenotype of microglia, suggesting a potential therapeutic target for neuroinflammation.</p>","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":"74 1","pages":"87"},"PeriodicalIF":4.8,"publicationDate":"2025-05-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144173613","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}