Mitochondrial dysfunction drives Rheumatoid Arthritis (RA) progression by disturbing energy metabolism and promoting inflammation. Additionally, the female predominance of RA highlights estrogen deficiency as an important contributor to disease development. The effect of estrogen in RA has been investigated; however, its specific effects on the mitochondrial proteome and function have yet to be studied. This study investigated the effects of 17-β estradiol (E2) on the mitochondrial proteome of patient-derived RA fibroblast-like synoviocytes (RA-FLS) using Sequential Window Acquisition of all Theoretical Mass Spectra (SWATH-MS) analysis, followed by an assessment of key mitochondrial functional parameters and in vitro validation. The results identified an upregulated expression of two mitochondrial proteins, Acyl-CoA dehydrogenase very long chain (ACADVL) and ATP synthase subunit O (ATP5O), after E2 treatment in RA-FLS. This was further validated by increased real-time ATP production and reduced glycolytic capacity, along with increased expression of proteins related to fatty acid β-oxidation. In addition, E2 influenced mitochondrial dynamics by modulating the fission-fusion balance, resulting in improved mitochondrial morphology. E2 treatment also reduced the expression of mitophagy markers and increased mitochondrial membrane potential, indicating improved mitochondrial function. It also lowered mitochondria-centered oxidative stress by upregulating mitochondrial antioxidant enzymes. Mitochondrial proteomics analysis thus, demonstrated that E2 has the potential to enhance mitochondrial energy metabolism and alleviate mitochondrial dysfunction in RA. These findings provide a foundation for further exploration of mitochondria-targeted therapeutic approaches in RA management.
{"title":"Mitochondrial proteomics reveals the impact of Estrogen in enhancing energy metabolism of patient-derived fibroblast-like synoviocytes in rheumatoid arthritis.","authors":"Swati Malik, Debolina Chakraborty, Prachi Agnihotri, Ashish Sarkar, Lovely Joshi, Mohd Saquib, Vijay Kumar, Sagarika Biswas","doi":"10.1007/s00011-025-02120-6","DOIUrl":"10.1007/s00011-025-02120-6","url":null,"abstract":"<p><p>Mitochondrial dysfunction drives Rheumatoid Arthritis (RA) progression by disturbing energy metabolism and promoting inflammation. Additionally, the female predominance of RA highlights estrogen deficiency as an important contributor to disease development. The effect of estrogen in RA has been investigated; however, its specific effects on the mitochondrial proteome and function have yet to be studied. This study investigated the effects of 17-β estradiol (E2) on the mitochondrial proteome of patient-derived RA fibroblast-like synoviocytes (RA-FLS) using Sequential Window Acquisition of all Theoretical Mass Spectra (SWATH-MS) analysis, followed by an assessment of key mitochondrial functional parameters and in vitro validation. The results identified an upregulated expression of two mitochondrial proteins, Acyl-CoA dehydrogenase very long chain (ACADVL) and ATP synthase subunit O (ATP5O), after E2 treatment in RA-FLS. This was further validated by increased real-time ATP production and reduced glycolytic capacity, along with increased expression of proteins related to fatty acid β-oxidation. In addition, E2 influenced mitochondrial dynamics by modulating the fission-fusion balance, resulting in improved mitochondrial morphology. E2 treatment also reduced the expression of mitophagy markers and increased mitochondrial membrane potential, indicating improved mitochondrial function. It also lowered mitochondria-centered oxidative stress by upregulating mitochondrial antioxidant enzymes. Mitochondrial proteomics analysis thus, demonstrated that E2 has the potential to enhance mitochondrial energy metabolism and alleviate mitochondrial dysfunction in RA. These findings provide a foundation for further exploration of mitochondria-targeted therapeutic approaches in RA management.</p>","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":"74 1","pages":"146"},"PeriodicalIF":5.4,"publicationDate":"2025-10-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145344894","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}
Objective: To investigate the expression of ferroptosis-related molecules in macrophages of community-acquired pneumonia (CAP) patients and explore their association with disease severity. This study provides novel insights into the role of ferroptosis in CAP pathogenesis by integrating transcriptome sequencing, RT-qPCR, Western blot, and flow cytometry analyses.
Methods: Ferroptosis-related molecules were analyzed in CD14⁺ monocytes and CD11b⁺ macrophages from CAP patients using transcriptome sequencing, RT-qPCR, Western blot, and flow cytometry. Clinical data from CAP patients (n = 46) and healthy controls (n = 63) were compared.
Results: CAP patients exhibited elevated levels of ferroptosis markers (PTGS2, Fe²⁺, and lipid peroxides) and downregulation of inhibitors (TP53 and GPX4). Transcriptome analysis showed activation of the ferroptosis pathway, with significant changes correlating with disease severity. Elevated neutrophil counts and decreased lymphocyte levels were also observed in CAP patients.
Conclusion: Ferroptosis is intricately involved in CAP pathogenesis, with altered expression of key molecules contributing to disease progression. Our findings highlight the potential of targeting ferroptosis-related molecules (e.g., TP53, GPX4, and PTGS2) as a novel therapeutic strategy to mitigate inflammation and tissue damage in CAP. Future studies should validate these findings in larger cohorts.
{"title":"Changes in ferroptosis-related molecular expression in macrophages of community-acquired pneumonia patients and their associated with the disease progression.","authors":"Huan-Shao Huang, Jia-Xin Chi, Le-Yao Xiao, Jia-Jun Wang, Shi-Ying Lai, Lan Chen, Jiang Pi, Yan-Guang Cong, Yi-Ming Shao, Jun-Fa Xu","doi":"10.1007/s00011-025-02116-2","DOIUrl":"10.1007/s00011-025-02116-2","url":null,"abstract":"<p><strong>Objective: </strong>To investigate the expression of ferroptosis-related molecules in macrophages of community-acquired pneumonia (CAP) patients and explore their association with disease severity. This study provides novel insights into the role of ferroptosis in CAP pathogenesis by integrating transcriptome sequencing, RT-qPCR, Western blot, and flow cytometry analyses.</p><p><strong>Methods: </strong>Ferroptosis-related molecules were analyzed in CD14⁺ monocytes and CD11b⁺ macrophages from CAP patients using transcriptome sequencing, RT-qPCR, Western blot, and flow cytometry. Clinical data from CAP patients (n = 46) and healthy controls (n = 63) were compared.</p><p><strong>Results: </strong>CAP patients exhibited elevated levels of ferroptosis markers (PTGS2, Fe²⁺, and lipid peroxides) and downregulation of inhibitors (TP53 and GPX4). Transcriptome analysis showed activation of the ferroptosis pathway, with significant changes correlating with disease severity. Elevated neutrophil counts and decreased lymphocyte levels were also observed in CAP patients.</p><p><strong>Conclusion: </strong>Ferroptosis is intricately involved in CAP pathogenesis, with altered expression of key molecules contributing to disease progression. Our findings highlight the potential of targeting ferroptosis-related molecules (e.g., TP53, GPX4, and PTGS2) as a novel therapeutic strategy to mitigate inflammation and tissue damage in CAP. Future studies should validate these findings in larger cohorts.</p>","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":"74 1","pages":"144"},"PeriodicalIF":5.4,"publicationDate":"2025-10-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12534296/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145312887","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}
Pub Date : 2025-10-17DOI: 10.1007/s00011-025-02110-8
Linghui Meng, Jing Liu, Hsiao Hui Ong, De-Yun Wang, Li Shi
Emerging evidence indicates that upper and lower airway diseases share anatomical and pathophysiological features. Infections often begin in the upper airway and progress downward, suggesting common immunological mechanisms. Mucin 1 (MUC1), a membrane-bound glycoprotein abundantly expressed in airway epithelial cells, has attracted increasing attention for its immunoregulatory and barrier functions. This review summarizes recent findings on MUC1's involvement in airway inflammation driven by Th1, Th2, and Th17 immune responses. In Th1-type inflammation, MUC1 negatively regulates Toll-like receptor (TLR)-NF-κB signaling pathways, thereby limiting excessive inflammatory responses to bacterial and viral infections. In Th2-type inflammation, MUC1 influences eosinophil survival, maintains epithelial integrity, and modulates glucocorticoid sensitivity, exerting both protective and pathological effects. In Th17-type inflammation, characterized by neutrophil infiltration and elevated IL-17A and IL-22, MUC1 expression alleviates chronic inflammation and may impact microbiome dysbiosis. While MUC1's roles in lower airway disorders are increasingly understood, its specific function and regulatory mechanisms in upper airway diseases remain unclear. This review adopts the unified airway disease (UAD) framework to examine the endotype-specific roles of MUC1 across the upper and lower airways. Rather than providing a disease-by-disease summary, we synthesize evidence through Th1/Th2/Th17 endotypes, link shared mechanisms to biomarker-based patient stratification, and outline MUC1-targeted therapeutic strategies. By applying an endotype- and UAD-centered perspective, the review distinguishes itself from previous work and highlights actionable opportunities for precision medicine. Furthermore, we emphasize the translational potential of MUC1 as both a diagnostic biomarker and a therapeutic target, focusing on advances in small peptides, monoclonal antibodies, RNA interference, and natural compounds that modulate MUC1-related pathways. These developments may ultimately enable the creation of personalized therapies for airway inflammation.
{"title":"MUC1 in the upper-lower airway inflammatory continuum: an endotype-centered perspective.","authors":"Linghui Meng, Jing Liu, Hsiao Hui Ong, De-Yun Wang, Li Shi","doi":"10.1007/s00011-025-02110-8","DOIUrl":"10.1007/s00011-025-02110-8","url":null,"abstract":"<p><p>Emerging evidence indicates that upper and lower airway diseases share anatomical and pathophysiological features. Infections often begin in the upper airway and progress downward, suggesting common immunological mechanisms. Mucin 1 (MUC1), a membrane-bound glycoprotein abundantly expressed in airway epithelial cells, has attracted increasing attention for its immunoregulatory and barrier functions. This review summarizes recent findings on MUC1's involvement in airway inflammation driven by Th1, Th2, and Th17 immune responses. In Th1-type inflammation, MUC1 negatively regulates Toll-like receptor (TLR)-NF-κB signaling pathways, thereby limiting excessive inflammatory responses to bacterial and viral infections. In Th2-type inflammation, MUC1 influences eosinophil survival, maintains epithelial integrity, and modulates glucocorticoid sensitivity, exerting both protective and pathological effects. In Th17-type inflammation, characterized by neutrophil infiltration and elevated IL-17A and IL-22, MUC1 expression alleviates chronic inflammation and may impact microbiome dysbiosis. While MUC1's roles in lower airway disorders are increasingly understood, its specific function and regulatory mechanisms in upper airway diseases remain unclear. This review adopts the unified airway disease (UAD) framework to examine the endotype-specific roles of MUC1 across the upper and lower airways. Rather than providing a disease-by-disease summary, we synthesize evidence through Th1/Th2/Th17 endotypes, link shared mechanisms to biomarker-based patient stratification, and outline MUC1-targeted therapeutic strategies. By applying an endotype- and UAD-centered perspective, the review distinguishes itself from previous work and highlights actionable opportunities for precision medicine. Furthermore, we emphasize the translational potential of MUC1 as both a diagnostic biomarker and a therapeutic target, focusing on advances in small peptides, monoclonal antibodies, RNA interference, and natural compounds that modulate MUC1-related pathways. These developments may ultimately enable the creation of personalized therapies for airway inflammation.</p>","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":"74 1","pages":"143"},"PeriodicalIF":5.4,"publicationDate":"2025-10-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145312857","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}
Pub Date : 2025-10-17DOI: 10.1007/s00011-025-02112-6
Víctor Serrano-Fernández, Juan Manuel Carmona-Torres, Ángel López-Fernández-Roldán, Matilde Isabel Castillo-Hermoso, Sergio Rodríguez-Cañamero, Rosa María Molina-Madueño, José Alberto Laredo-Aguilera
Background and objective: Inflammatory bowel diseases, including Crohn's disease and ulcerative colitis (UC), are chronic conditions characterized by intestinal inflammation. Soluble fiber is fermented by gut microbiota into short-chain fatty acids (SCFA), which possess anti-inflammatory properties. This review aimed to assess the efficacy of oral and topical SCFA administration in patients with UC.
Methods: A systematic review with meta-analysis was conducted in accordance with PRISMA guidelines. Meta-analyses were performed using means and standard deviations to assess clinical and histological outcomes. Endoscopic criteria were also evaluated to determine the effectiveness of the intervention.
Results: Nine studies compared SCFA supplementation with standard therapy, and one employed a crossover design. Oral SCFA, when combined with standard treatment, as associated with reductions in fecal calprotectin and C-reactive protein levels. Meta-analyses of topical SCFA administration revealed a standardized mean difference of - 0.29 ± 0.22, (95% CI: - 0.65 to 0.07; heterogeneity I2 = 28%) for the Ulcerative Colitis Disease Activity Index score, and - 0.73 ± 0.61, (95% CI: - 1.58, 0.12; heterogeneity I2 = 64%) for histological scores. Endoscopic scores decreased in both intervention and control groups. No adverse effects were observed.
Conclusion: SCFA administration, either orally or topically, has been investigated as a potential adjunct to standard UC therapies. However, the current evidence is limited, particularly for oral administration, which has only been assessed in two studies. Further research is needed to clarify the potential role of SCFA administration.
{"title":"Short-chain fatty acids in the treatment of ulcerative colitis. Systematic review and meta-analysis.","authors":"Víctor Serrano-Fernández, Juan Manuel Carmona-Torres, Ángel López-Fernández-Roldán, Matilde Isabel Castillo-Hermoso, Sergio Rodríguez-Cañamero, Rosa María Molina-Madueño, José Alberto Laredo-Aguilera","doi":"10.1007/s00011-025-02112-6","DOIUrl":"10.1007/s00011-025-02112-6","url":null,"abstract":"<p><strong>Background and objective: </strong>Inflammatory bowel diseases, including Crohn's disease and ulcerative colitis (UC), are chronic conditions characterized by intestinal inflammation. Soluble fiber is fermented by gut microbiota into short-chain fatty acids (SCFA), which possess anti-inflammatory properties. This review aimed to assess the efficacy of oral and topical SCFA administration in patients with UC.</p><p><strong>Methods: </strong>A systematic review with meta-analysis was conducted in accordance with PRISMA guidelines. Meta-analyses were performed using means and standard deviations to assess clinical and histological outcomes. Endoscopic criteria were also evaluated to determine the effectiveness of the intervention.</p><p><strong>Results: </strong>Nine studies compared SCFA supplementation with standard therapy, and one employed a crossover design. Oral SCFA, when combined with standard treatment, as associated with reductions in fecal calprotectin and C-reactive protein levels. Meta-analyses of topical SCFA administration revealed a standardized mean difference of - 0.29 ± 0.22, (95% CI: - 0.65 to 0.07; heterogeneity I<sup>2</sup> = 28%) for the Ulcerative Colitis Disease Activity Index score, and - 0.73 ± 0.61, (95% CI: - 1.58, 0.12; heterogeneity I<sup>2</sup> = 64%) for histological scores. Endoscopic scores decreased in both intervention and control groups. No adverse effects were observed.</p><p><strong>Conclusion: </strong>SCFA administration, either orally or topically, has been investigated as a potential adjunct to standard UC therapies. However, the current evidence is limited, particularly for oral administration, which has only been assessed in two studies. Further research is needed to clarify the potential role of SCFA administration.</p>","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":"74 1","pages":"142"},"PeriodicalIF":5.4,"publicationDate":"2025-10-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145312896","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}
<p><strong>Background: </strong>Previous studies have indicated a correlation between the glycosylated hemoglobin index (HGI) and the prognosis of patients with sepsis. However, the impact of its dynamic fluctuations on patient outcomes remains insufficiently explored. This study seeks to investigate the relationship between HGI trajectory changes over time and the prognosis of patients with sepsis, and developing an optimal predictive model for 28-day and 90-day mortality risk using machine learning techniques.</p><p><strong>Methods: </strong>Data from the MIMIC-IV 3.0 database were employed to construct a linear regression model utilizing glycosylated hemoglobin (HbA1c) and fasting blood glucose (FBG) measured at six distinct time points to calculate HGI. Trajectory analysis revealed three distinct HGI change patterns: rising (RS), stable (ST), and descending (DS). Based on these groupings, Kaplan-Meier survival curves, Cox regression models, and mediation analysis were applied. Lasso regression was utilized for feature selection, and four machine learning models-Extreme Gradient Boosting (XGBoost), Support Vector Machine (SVM), Logistic Regression, and Random Forest (RF)-were developed and evaluated through ROC curves, Decision Curve Analysis (DCA), and calibration curves. SHapley Additive exPlanations (SHAP) values were incorporated for interpretability, and nomograms for 28-day and 90-day mortality were generated.</p><p><strong>Results: </strong>A total of 2,616 patients were included in the analysis, with 407 patients (15.56%) dying within 28 days. multivariable Cox regression analysis, using the ST group as a reference, revealed that patients in the RS group had a significantly higher risk of death at both 28 days and 90 days, whereas those in the DS group demonstrated a markedly reduced risk. These findings were consistent across all models. Among the models evaluated, the Logistic Regression model exhibited the highest predictive accuracy, with AUC values for the 28-day mortality prediction in the training and validation sets of 0.743 and 0.732, respectively, and for 90-day mortality of 0.748 and 0.735. The key predictive factors for 28-day mortality included albumin, HGI, renal injury, and the APSIII score, while for 90-day mortality, renal injury, APSIII score, HGI, and albumin were the most influential variables. To enhance the clinical applicability of these models, nomograms for 28-day and 90-day mortality were constructed, achieving AUCs of 0.715 and 0.747, respectively. Calibration curves demonstrated strong concordance between predicted probabilities and observed outcomes.</p><p><strong>Conclusion: </strong>Dynamic alterations in HGI during follow-up were found to be significantly associated with the risk of mortality in patients with sepsis at both 28 days and 90 days. This study presents a machine learning-based model for predicting the mortality risk of patients with sepsis, with potential clinical utility in early risk strati
{"title":"Dynamic HGI trajectories and their impact on survival in patients with sepsis: a machine learning prognostic model.","authors":"Aifeng He, Wei Jiang, Jing Fu, Leiming Xu, Congliang You, Suhui Li, Jiangquan Yu, Ruiqiang Zheng","doi":"10.1007/s00011-025-02113-5","DOIUrl":"10.1007/s00011-025-02113-5","url":null,"abstract":"<p><strong>Background: </strong>Previous studies have indicated a correlation between the glycosylated hemoglobin index (HGI) and the prognosis of patients with sepsis. However, the impact of its dynamic fluctuations on patient outcomes remains insufficiently explored. This study seeks to investigate the relationship between HGI trajectory changes over time and the prognosis of patients with sepsis, and developing an optimal predictive model for 28-day and 90-day mortality risk using machine learning techniques.</p><p><strong>Methods: </strong>Data from the MIMIC-IV 3.0 database were employed to construct a linear regression model utilizing glycosylated hemoglobin (HbA1c) and fasting blood glucose (FBG) measured at six distinct time points to calculate HGI. Trajectory analysis revealed three distinct HGI change patterns: rising (RS), stable (ST), and descending (DS). Based on these groupings, Kaplan-Meier survival curves, Cox regression models, and mediation analysis were applied. Lasso regression was utilized for feature selection, and four machine learning models-Extreme Gradient Boosting (XGBoost), Support Vector Machine (SVM), Logistic Regression, and Random Forest (RF)-were developed and evaluated through ROC curves, Decision Curve Analysis (DCA), and calibration curves. SHapley Additive exPlanations (SHAP) values were incorporated for interpretability, and nomograms for 28-day and 90-day mortality were generated.</p><p><strong>Results: </strong>A total of 2,616 patients were included in the analysis, with 407 patients (15.56%) dying within 28 days. multivariable Cox regression analysis, using the ST group as a reference, revealed that patients in the RS group had a significantly higher risk of death at both 28 days and 90 days, whereas those in the DS group demonstrated a markedly reduced risk. These findings were consistent across all models. Among the models evaluated, the Logistic Regression model exhibited the highest predictive accuracy, with AUC values for the 28-day mortality prediction in the training and validation sets of 0.743 and 0.732, respectively, and for 90-day mortality of 0.748 and 0.735. The key predictive factors for 28-day mortality included albumin, HGI, renal injury, and the APSIII score, while for 90-day mortality, renal injury, APSIII score, HGI, and albumin were the most influential variables. To enhance the clinical applicability of these models, nomograms for 28-day and 90-day mortality were constructed, achieving AUCs of 0.715 and 0.747, respectively. Calibration curves demonstrated strong concordance between predicted probabilities and observed outcomes.</p><p><strong>Conclusion: </strong>Dynamic alterations in HGI during follow-up were found to be significantly associated with the risk of mortality in patients with sepsis at both 28 days and 90 days. This study presents a machine learning-based model for predicting the mortality risk of patients with sepsis, with potential clinical utility in early risk strati","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":"74 1","pages":"145"},"PeriodicalIF":5.4,"publicationDate":"2025-10-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145312829","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}
Interleukin-15 (IL-15) is a pleiotropic cytokine of the common γ-chain (γc) family, playing a central role in the development, survival, and activation of natural killer (NK) cells and memory CD8+ T cells. Its tightly regulated expression is essential for maintaining immune homeostasis, while dysregulation of IL-15 signaling has been implicated in the pathogenesis of various autoimmune and chronic inflammatory diseases. This review explores the structural features, receptor complex, and signaling mechanisms of IL-15, emphasizing its immunomodulatory functions in both innate and adaptive immunity. We highlight the role of IL-15 in the initiation and progression of autoimmune conditions such as rheumatoid arthritis, systemic lupus erythematosus, multiple sclerosis (MS), type 1 diabetes, celiac disease, inflammatory bowel disease, psoriasis, and ankylosing spondylitis. Special focus is given to IL-15-mediated activation of tissue-resident memory T cells (TRM), cytotoxic CD8+ T cells, and NK cells, which contribute to tissue-specific autoimmunity and sustained inflammation. Furthermore, we summarize emerging therapeutic strategies targeting IL-15 and its receptor complex, including monoclonal antibodies, receptor antagonists, and cytokine fusion proteins. Clinical trials and preclinical data suggest that IL-15 blockade may offer a promising therapeutic avenue in autoimmune diseases, particularly in refractory or relapsing forms. The review concludes with future directions for integrating IL-15-targeted interventions into precision immunotherapy.
{"title":"Interleukin 15 and autoimmune disorders: pathophysiology, therapeutic potential, and clinical implications.","authors":"Leila Darouni, Fatemeh Tavassoli Razavi, Esmaeil Yazdanpanah, Niloufar Orooji, Alireza Shadab, Atena Emami, Haniyeh Molavi, Dariush Haghmorad","doi":"10.1007/s00011-025-02084-7","DOIUrl":"10.1007/s00011-025-02084-7","url":null,"abstract":"<p><p>Interleukin-15 (IL-15) is a pleiotropic cytokine of the common γ-chain (γc) family, playing a central role in the development, survival, and activation of natural killer (NK) cells and memory CD8<sup>+</sup> T cells. Its tightly regulated expression is essential for maintaining immune homeostasis, while dysregulation of IL-15 signaling has been implicated in the pathogenesis of various autoimmune and chronic inflammatory diseases. This review explores the structural features, receptor complex, and signaling mechanisms of IL-15, emphasizing its immunomodulatory functions in both innate and adaptive immunity. We highlight the role of IL-15 in the initiation and progression of autoimmune conditions such as rheumatoid arthritis, systemic lupus erythematosus, multiple sclerosis (MS), type 1 diabetes, celiac disease, inflammatory bowel disease, psoriasis, and ankylosing spondylitis. Special focus is given to IL-15-mediated activation of tissue-resident memory T cells (TRM), cytotoxic CD8<sup>+</sup> T cells, and NK cells, which contribute to tissue-specific autoimmunity and sustained inflammation. Furthermore, we summarize emerging therapeutic strategies targeting IL-15 and its receptor complex, including monoclonal antibodies, receptor antagonists, and cytokine fusion proteins. Clinical trials and preclinical data suggest that IL-15 blockade may offer a promising therapeutic avenue in autoimmune diseases, particularly in refractory or relapsing forms. The review concludes with future directions for integrating IL-15-targeted interventions into precision immunotherapy.</p>","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":"74 1","pages":"141"},"PeriodicalIF":5.4,"publicationDate":"2025-10-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145312824","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}
Pub Date : 2025-10-03DOI: 10.1007/s00011-025-02105-5
Shuyu Wang, Hongquan Jiang, Ting Yao
Objective: This multiomics study investigated causal relationships between the gut microbiota (GM), immune dysregulation, and amyotrophic lateral sclerosis (ALS) pathogenesis using Mendelian randomization (MR) with experimental validation.
Materials: Analyses incorporated genome-wide data from 87,347 participants (GM: n = 7738; ALS: 20,806 patients, 59,804 controls; immune phenotypes: n = 3757), transcriptomic data from 71 subjects (56 ALS patients, 15 controls), and experimental validation in matched cohorts (n = 6 subjects per group).
Methods: Two-sample bidirectional MR and mediation analysis were used to evaluate associations. Experimental validation employed flow cytometry for myeloid-derived suppressor cell quantification, enzyme-linked immunosorbent assay for cytokines, and real-time polymerase chain reaction for bacterial validation. Statistical analyses included inverse variance weighted methods with Cohen's d calculations.
Results: Sixteen bacterial taxa, including p_Firmicutes.c_Clostridia, displayed protective associations with the risk of ALS, whereas sixteen showed harmful associations. Mediation analysis suggested that p_Firmicutes.c_Clostridia may confer protection through CD33+HLA-DR-myeloid-derived suppressor cell upregulation (23.8% mediation effect). Experimental validation confirmed fewer myeloid-derived suppressor cells in ALS patients (4.0 ± 0.8% vs. 7.5 ± 1.0%, p < 0.001, Cohen's d = 1.4) and lower levels of anti-inflammatory cytokines (TGF-β1: Cohen's d = 1.8, p < 0.001).
Conclusions: These findings support causal associations between gut microbial taxa and the ALS risk, which are mediated through immunoregulatory mechanisms, highlighting therapeutic targets within the gut‒immune‒brain axis.
目的:本多组学研究利用孟德尔随机化(MR)研究了肠道微生物群(GM)、免疫失调和肌萎缩性侧索硬化症(ALS)发病机制之间的因果关系,并进行了实验验证。材料:分析纳入了来自87,347名参与者的全基因组数据(GM: n = 7738; ALS: 20,806名患者,59,804名对照;免疫表型:n = 3757),来自71名受试者(56名ALS患者,15名对照)的转录组数据,以及匹配队列(每组n = 6名受试者)的实验验证。方法:采用双样本双向磁共振和中介分析来评估相关性。实验验证采用流式细胞术进行髓源性抑制细胞定量,酶联免疫吸附测定细胞因子,实时聚合酶链反应进行细菌验证。统计分析包括用Cohen's d计算的逆方差加权方法。结果:16个细菌分类群,包括p_firmicutes、c_clostridia,与ALS的发病风险呈保护性相关,16个细菌分类群与ALS的发病风险呈有害相关。调解分析表明,p_Firmicutes.c_Clostridia可能通过上调CD33+ hla - dr -髓源性抑制细胞来发挥保护作用(调解作用为23.8%)。实验验证证实,ALS患者骨髓源性抑制细胞较少(4.0±0.8% vs. 7.5±1.0%)。结论:这些发现支持肠道微生物分类群与ALS风险之间的因果关系,这是通过免疫调节机制介导的,突出了肠道-免疫-脑轴内的治疗靶点。
{"title":"Integrative genomics and functional immunology reveal Clostridia species as modulators of neuroinflammation in amyotrophic lateral sclerosis.","authors":"Shuyu Wang, Hongquan Jiang, Ting Yao","doi":"10.1007/s00011-025-02105-5","DOIUrl":"10.1007/s00011-025-02105-5","url":null,"abstract":"<p><strong>Objective: </strong>This multiomics study investigated causal relationships between the gut microbiota (GM), immune dysregulation, and amyotrophic lateral sclerosis (ALS) pathogenesis using Mendelian randomization (MR) with experimental validation.</p><p><strong>Materials: </strong>Analyses incorporated genome-wide data from 87,347 participants (GM: n = 7738; ALS: 20,806 patients, 59,804 controls; immune phenotypes: n = 3757), transcriptomic data from 71 subjects (56 ALS patients, 15 controls), and experimental validation in matched cohorts (n = 6 subjects per group).</p><p><strong>Methods: </strong>Two-sample bidirectional MR and mediation analysis were used to evaluate associations. Experimental validation employed flow cytometry for myeloid-derived suppressor cell quantification, enzyme-linked immunosorbent assay for cytokines, and real-time polymerase chain reaction for bacterial validation. Statistical analyses included inverse variance weighted methods with Cohen's d calculations.</p><p><strong>Results: </strong>Sixteen bacterial taxa, including p_Firmicutes.c_Clostridia, displayed protective associations with the risk of ALS, whereas sixteen showed harmful associations. Mediation analysis suggested that p_Firmicutes.c_Clostridia may confer protection through CD33+HLA-DR-myeloid-derived suppressor cell upregulation (23.8% mediation effect). Experimental validation confirmed fewer myeloid-derived suppressor cells in ALS patients (4.0 ± 0.8% vs. 7.5 ± 1.0%, p < 0.001, Cohen's d = 1.4) and lower levels of anti-inflammatory cytokines (TGF-β1: Cohen's d = 1.8, p < 0.001).</p><p><strong>Conclusions: </strong>These findings support causal associations between gut microbial taxa and the ALS risk, which are mediated through immunoregulatory mechanisms, highlighting therapeutic targets within the gut‒immune‒brain axis.</p>","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":"74 1","pages":"136"},"PeriodicalIF":5.4,"publicationDate":"2025-10-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145212665","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}
Pub Date : 2025-10-03DOI: 10.1007/s00011-025-02102-8
Jia-Yi Zhang, Qiu-Ni Su, Han Lin, Wei Lin, Mao-Lin Cui, Zhuo-Ying Huang, Bei-Ning Ye, Ying-Xin Ye, Yi-Lin Jia, Qing-Ying Zhu, Zhi-Han Li, Min-Ting Lin, Ning Wang, Bing-Long Wang, Shi-Rui Gan
Background: Neuroinflammation plays a recognized role in the pathogenesis of Spinocerebellar Ataxia Type 3 (SCA3). However, the involvement of systemic inflammatory responses in SCA3 remains poorly defined.
Objectives: Our study aimed to characterize peripheral inflammation in patients with SCA3, examine the relationship between peripheral inflammatory biomarkers and clinical/genetic features, and evaluate the diagnostic utility of these markers.
Methods: The cross-sectional study enrolled 101 patients with SCA3 and 101 healthy controls (HCs). The differences in peripheral inflammatory markers between patients with SCA3 and HCs were assessed. Multivariate linear regressions were used to analyze the associations between blood cell count-derived indices, C-reactive protein (CRP), and clinical/genetic features of SCA3. ROC curves were conducted to assess the potential of these markers to distinguish patients with SCA3 from HCs.
Results: Compared to HCs, patients with SCA3 exhibited significantly higher levels of leukocytes, neutrophils, monocytes, platelets, neutrophil-to-lymphocyte ratio (NLR), monocyte-to-lymphocyte ratio (MLR), systemic inflammatory index (SII), systemic inflammatory composite index (AISI), and CRP (adj. p < 0.05). Age influenced lgCRP, NLR, MLR, and lg(neutrophil-to-platelet ratio) (p < 0.05). CAG affected MLR and AISI (p < 0.05). The combination of BMI, monocytes, NLR, and SII differentiated patients with SCA3 from HCs (AUC = 0.779).
Conclusion: Patients with SCA3 display distinct peripheral inflammatory profiles, which correlate with clinical/genetic factors. These peripheral inflammatory markers hold promise as potential tools for diagnosing and monitoring SCA3.
{"title":"Peripheral inflammation in spinocerebellar ataxia type 3: associations with genetic and clinical manifestations.","authors":"Jia-Yi Zhang, Qiu-Ni Su, Han Lin, Wei Lin, Mao-Lin Cui, Zhuo-Ying Huang, Bei-Ning Ye, Ying-Xin Ye, Yi-Lin Jia, Qing-Ying Zhu, Zhi-Han Li, Min-Ting Lin, Ning Wang, Bing-Long Wang, Shi-Rui Gan","doi":"10.1007/s00011-025-02102-8","DOIUrl":"10.1007/s00011-025-02102-8","url":null,"abstract":"<p><strong>Background: </strong>Neuroinflammation plays a recognized role in the pathogenesis of Spinocerebellar Ataxia Type 3 (SCA3). However, the involvement of systemic inflammatory responses in SCA3 remains poorly defined.</p><p><strong>Objectives: </strong>Our study aimed to characterize peripheral inflammation in patients with SCA3, examine the relationship between peripheral inflammatory biomarkers and clinical/genetic features, and evaluate the diagnostic utility of these markers.</p><p><strong>Methods: </strong>The cross-sectional study enrolled 101 patients with SCA3 and 101 healthy controls (HCs). The differences in peripheral inflammatory markers between patients with SCA3 and HCs were assessed. Multivariate linear regressions were used to analyze the associations between blood cell count-derived indices, C-reactive protein (CRP), and clinical/genetic features of SCA3. ROC curves were conducted to assess the potential of these markers to distinguish patients with SCA3 from HCs.</p><p><strong>Results: </strong>Compared to HCs, patients with SCA3 exhibited significantly higher levels of leukocytes, neutrophils, monocytes, platelets, neutrophil-to-lymphocyte ratio (NLR), monocyte-to-lymphocyte ratio (MLR), systemic inflammatory index (SII), systemic inflammatory composite index (AISI), and CRP (adj. p < 0.05). Age influenced lgCRP, NLR, MLR, and lg(neutrophil-to-platelet ratio) (p < 0.05). CAG affected MLR and AISI (p < 0.05). The combination of BMI, monocytes, NLR, and SII differentiated patients with SCA3 from HCs (AUC = 0.779).</p><p><strong>Conclusion: </strong>Patients with SCA3 display distinct peripheral inflammatory profiles, which correlate with clinical/genetic factors. These peripheral inflammatory markers hold promise as potential tools for diagnosing and monitoring SCA3.</p>","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":"74 1","pages":"137"},"PeriodicalIF":5.4,"publicationDate":"2025-10-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145212668","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}
Pub Date : 2025-10-03DOI: 10.1007/s00011-025-02107-3
Qingxiang Zeng, Xiangqian Qiu, Yinhui Zeng, Xi Luo, Jing Ma, Wenlong Liu
Background: Apolipoprotein E (ApoE) promoted neutrophilic airway inflammation in mice with allergic asthma. Although group 2 innate lymphoid cells (ILC2s) have been established as pivotal mediators in allergic rhinitis (AR) pathogenesis, endogenous mechanisms regulating their hyperactivity are undefined. Whether ApoE has effects in ILC2s in AR is still unknown.
Objective: The aim of this investigation is to identify the crucial function of the suppressor modulator ApoE in regulating ILC2-mediated allergic airway inflammation.
Methods: A cohort comprising 15 pediatric AR patients and matched healthy controls was enrolled to assess serum ApoE messenger RNA (mRNA) expression and protein levels and their relationship with interleukin (IL)-5, IL-13 and total nasal symptoms scores (TNSS). In vitro experiments employing flow cytometry and enzyme-linked immunosorbent assay (ELISA) validated the regulatory effects of ApoE on ILC2s expansion capacity and cytokine secretion. Transcript levels of GATA binding protein 3 (GATA3) and retinoid acid receptor related orphan receptor alpha (RORα) was examined using quantitative reverse transcription polymerase chain reaction (qRT-RCR). The changes of IL-25, IL-33 and thymic stromal lymphopoietin (TSLP) levels after stimulation of human nasal epithelial cells (HNECs) by Dermatophagoides pteronyssinus (Der p), ApoE and anti-Low-Density Lipoprotein Receptor (LDLR) were determined by ELISA. The effects of ApoE and anti-LDLR in vivo were using the ovalbumin (OVA)-induced murine model.
Results: Clinical analyses indicated elevated serum ApoE mRNA and protein levels in AR patients, positively correlating with TNSS. Recombinant ApoE effectively promoted ILC2 activation in PBMCs from AR patients, inducing GATA3 and RORα expression and IL-5, IL-13 secretion, while these effects of ApoE were eliminated by anti-LDLR. Mechanistically, ApoE augmented the epithelial-ILC2 crosstalk by inducing nasal epithelial TSLP/IL-25/IL-33 release. Additionally, our studies have revealed that the anti-LDLR effectively reversed the effects of ApoE on those cytokine production. In vivo, administering anti-LDLR to AR mice effectively alleviates OVA and ApoE induced allergic airway inflammation, reducing eosinophilic infiltration and nasal symptoms, as well as OVA specific immunoglobulin E (IgE) and ILC2 proliferation.
Conclusions: As our findings demonstrate, targeting the ApoE-LDLR axis has considerable therapeutic promise in treating ILC2-dependent allergic airway inflammation.
{"title":"Apolipoprotein E promoted the proliferation and function of group 2 innate lymphoid cells through low density lipoprotein receptor in allergic rhinitis.","authors":"Qingxiang Zeng, Xiangqian Qiu, Yinhui Zeng, Xi Luo, Jing Ma, Wenlong Liu","doi":"10.1007/s00011-025-02107-3","DOIUrl":"10.1007/s00011-025-02107-3","url":null,"abstract":"<p><strong>Background: </strong>Apolipoprotein E (ApoE) promoted neutrophilic airway inflammation in mice with allergic asthma. Although group 2 innate lymphoid cells (ILC2s) have been established as pivotal mediators in allergic rhinitis (AR) pathogenesis, endogenous mechanisms regulating their hyperactivity are undefined. Whether ApoE has effects in ILC2s in AR is still unknown.</p><p><strong>Objective: </strong>The aim of this investigation is to identify the crucial function of the suppressor modulator ApoE in regulating ILC2-mediated allergic airway inflammation.</p><p><strong>Methods: </strong>A cohort comprising 15 pediatric AR patients and matched healthy controls was enrolled to assess serum ApoE messenger RNA (mRNA) expression and protein levels and their relationship with interleukin (IL)-5, IL-13 and total nasal symptoms scores (TNSS). In vitro experiments employing flow cytometry and enzyme-linked immunosorbent assay (ELISA) validated the regulatory effects of ApoE on ILC2s expansion capacity and cytokine secretion. Transcript levels of GATA binding protein 3 (GATA3) and retinoid acid receptor related orphan receptor alpha (RORα) was examined using quantitative reverse transcription polymerase chain reaction (qRT-RCR). The changes of IL-25, IL-33 and thymic stromal lymphopoietin (TSLP) levels after stimulation of human nasal epithelial cells (HNECs) by Dermatophagoides pteronyssinus (Der p), ApoE and anti-Low-Density Lipoprotein Receptor (LDLR) were determined by ELISA. The effects of ApoE and anti-LDLR in vivo were using the ovalbumin (OVA)-induced murine model.</p><p><strong>Results: </strong>Clinical analyses indicated elevated serum ApoE mRNA and protein levels in AR patients, positively correlating with TNSS. Recombinant ApoE effectively promoted ILC2 activation in PBMCs from AR patients, inducing GATA3 and RORα expression and IL-5, IL-13 secretion, while these effects of ApoE were eliminated by anti-LDLR. Mechanistically, ApoE augmented the epithelial-ILC2 crosstalk by inducing nasal epithelial TSLP/IL-25/IL-33 release. Additionally, our studies have revealed that the anti-LDLR effectively reversed the effects of ApoE on those cytokine production. In vivo, administering anti-LDLR to AR mice effectively alleviates OVA and ApoE induced allergic airway inflammation, reducing eosinophilic infiltration and nasal symptoms, as well as OVA specific immunoglobulin E (IgE) and ILC2 proliferation.</p><p><strong>Conclusions: </strong>As our findings demonstrate, targeting the ApoE-LDLR axis has considerable therapeutic promise in treating ILC2-dependent allergic airway inflammation.</p>","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":"74 1","pages":"139"},"PeriodicalIF":5.4,"publicationDate":"2025-10-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145212639","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}
Pub Date : 2025-10-03DOI: 10.1007/s00011-025-02098-1
Aishat Azeez, John A Baugh
Background: Lung disease remains a leading cause of global morbidity and mortality, with prevalence strongly influenced by lifestyle factors, including dietary patterns such as the Western diet. Chronic lung inflammation, driven by dysregulated immune responses, is a hallmark of many pulmonary conditions and exacerbates disease progression and severity Emerging evidence highlights potentially critical role of for Dietary fibre and it's metabolites particularly short chain fatty acids (SCFAs), acetate, butyrate and propionate, in modulating the gut-lung axis and regulating pulmonary immune response.
Objective: This review summarizes current evidence on how dietary fibre and SCFAs influence pulmonary immunity and inflammation through systemic and local mechanisms.MethodsLiterature on dietary fibre intake, SCFA production, and immune regulation in the context of lung disease was reviewed to identify key effects and mechanistic insights.
Findings: SCFAs, including acetate, butyrate, and propionate, are produced by gut microbial fermentation of fibre and act via G-protein coupled receptor signalling and histone deacetylase inhibition. These metabolites modulate epithelial and immune cell function, reduce inflammation, and enhance lung immune protection. Beyond local effects, SCFAs influence hematopoietic cells in the bone marrow, altering their recruitment and activity in the lung.
Conclusions: Dietary fibre intake and SCFA-mediated gut-lung immune regulation represent a promising area for therapeutic development. A deeper understanding of these pathways may support novel strategies for the prevention and treatment of respiratory diseases.
{"title":"The role of dietary fibre in lung inflammation: microbiota, metabolites, and immune crosstalk.","authors":"Aishat Azeez, John A Baugh","doi":"10.1007/s00011-025-02098-1","DOIUrl":"10.1007/s00011-025-02098-1","url":null,"abstract":"<p><strong>Background: </strong>Lung disease remains a leading cause of global morbidity and mortality, with prevalence strongly influenced by lifestyle factors, including dietary patterns such as the Western diet. Chronic lung inflammation, driven by dysregulated immune responses, is a hallmark of many pulmonary conditions and exacerbates disease progression and severity Emerging evidence highlights potentially critical role of for Dietary fibre and it's metabolites particularly short chain fatty acids (SCFAs), acetate, butyrate and propionate, in modulating the gut-lung axis and regulating pulmonary immune response.</p><p><strong>Objective: </strong>This review summarizes current evidence on how dietary fibre and SCFAs influence pulmonary immunity and inflammation through systemic and local mechanisms.MethodsLiterature on dietary fibre intake, SCFA production, and immune regulation in the context of lung disease was reviewed to identify key effects and mechanistic insights.</p><p><strong>Findings: </strong>SCFAs, including acetate, butyrate, and propionate, are produced by gut microbial fermentation of fibre and act via G-protein coupled receptor signalling and histone deacetylase inhibition. These metabolites modulate epithelial and immune cell function, reduce inflammation, and enhance lung immune protection. Beyond local effects, SCFAs influence hematopoietic cells in the bone marrow, altering their recruitment and activity in the lung.</p><p><strong>Conclusions: </strong>Dietary fibre intake and SCFA-mediated gut-lung immune regulation represent a promising area for therapeutic development. A deeper understanding of these pathways may support novel strategies for the prevention and treatment of respiratory diseases.</p>","PeriodicalId":13550,"journal":{"name":"Inflammation Research","volume":"74 1","pages":"135"},"PeriodicalIF":5.4,"publicationDate":"2025-10-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145212595","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}