{"title":"CAFs-released exosomal CREB1 promotes cell progression and immune evasion in thyroid cancer via the positive regulation of CCL20.","authors":"Chen Zheng, Hu Hei, Yifei Zhai, Wenbo Gong, Runfang Zhang, Songtao Zhang","doi":"10.1080/08916934.2025.2458324","DOIUrl":"https://doi.org/10.1080/08916934.2025.2458324","url":null,"abstract":"<p><strong>Background: </strong>Exosomes derived from cancer-associated fibroblasts (CAFs) can affect tumor microenvironment (TME) of thyroid cancer (TC). The cAMP response element binding protein 1 (CREB1) acts as a transcription factor to participate in cancer development. Currently, we aimed to explore the molecular mechanism of exosome-associated CREB1 and C-C motif chemokine ligand 20 (CCL20) in TC.</p><p><strong>Methods: </strong>The mRNA and protein levels were examined <i>via</i> RT-qPCR and western blot. Gene interaction was analyzed using ChIP and dual-luciferase reporter assays. Cell migration, invasion and proliferation were assessed by wound healing, transwell and EdU assays. Exosomes were characterized by morphology observation and western blot. The proliferation and apoptosis of CD8<sup>+</sup> T cells were detected by immunofluorescence and flow cytometry. <i>In vivo</i> assays were performed by establishing xenograft models.</p><p><strong>Results: </strong>CREB1 was highly expressed in TC. CREB1 positively interacted with CCL20 in TC. CREB1 facilitated TC cell migration, invasion and proliferation <i>via</i> targeting CCL20. CCL20 expression was reduced by transferring CAFs-secreted exosomes sheltering CREB1 downregulation. Exosomal CREB1 knockdown receded cell progression and enhanced CD8<sup>+</sup> T function by mediating CCL20. CAFs-associated exosomal CREB1 downregulation inhibited tumorigenesis through affecting CCL20.</p><p><strong>Conclusion: </strong>CAFs-derived exosomes accelerated the malignant behaviors and immune evasion in TC by carrying CREB1 to up-regulate CCL20.</p>","PeriodicalId":8688,"journal":{"name":"Autoimmunity","volume":"58 1","pages":"2458324"},"PeriodicalIF":3.3,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143035471","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Deciphering distinct pathogenic mechanisms of ankylosing spondylitis and systemic sclerosis via shared biomarkers ZSWIM6 and CCL3L3: Insights from an integrative bioinformatics approach.","authors":"Liangyu Huang, Jiarui Chen, Renbang Yang, Jianjun Shi, Chenxing Zhou, Tianyou Chen, Sitan Feng, Chengqian Huang, Jieping Huang, Jiang Xue, Zhongxian Zhou, Jichong Zhu, Shaofeng Wu, Xinli Zhan, Chong Liu","doi":"10.1080/08916934.2024.2445557","DOIUrl":"https://doi.org/10.1080/08916934.2024.2445557","url":null,"abstract":"<p><p>Ankylosing Spondylitis (AS) and Systemic Sclerosis (SSc) are both autoimmune diseases, albeit with distinct anatomical targets. AS primarily affects the spine and sacroiliac joints, triggering inflammation and eventual fusion of the vertebrae. SSc predominantly impacts the skin and connective tissues, leading to skin fibrosis, thickening, and potential damage to vital organs such as the lungs, heart, and kidneys. Despite their differing anatomical manifestations, inflammation serves as a pivotal factor in both conditions. Exploring the causes of the different pathogenesis of inflammation in AS and SSc could provide new insights into their treatment. We selected RNA-seq profiles of peripheral blood mononuclear cells (PBMCs) from the GEO datasets GSE73754 and GSE19617. DEGs were identified using the Limma R package with an adjusted <i>p</i>-value cutoff of < 0.05. Gene Ontology pathway analysis, SVM recursive feature elimination, and Gene Set Enrichment Analysis (GSEA) were conducted to analyze the DEGs. CIBERSORT was applied to estimate immune cell composition and its correlation with hub genes. Single-cell RNA sequencing data from peripheral blood mononuclear cells in the GSE194315 dataset were included to support differential expression analysis and biomarker identification. Additionally, single-cell RNA sequencing data from bone marrow blood samples were utilized to further validate these findings, offering complementary insights into biomarker expression across distinct sample types. A total of 762 DEGs were identified between AS patients and controls, and 441 DEGs between SSc patients and controls. Both conditions showed enrichment in the Natural killer cell mediated cytotoxicity pathway. ZSWIM6 and CCL3L3 were identified as potential biomarkers in AS and SSc, with significant diagnostic capabilities demonstrated by ROC analysis. Correlation analysis revealed associations between these biomarkers and specific immune cell types. The study utilizing ZSWIM6 and CCL3L3 as potential biomarkers provides deep insights into the distinct molecular mechanisms of SSc and AS. These findings lay the foundation for future research on targeted therapies and enhance our understanding of immune cell interactions in these autoimmune diseases.</p>","PeriodicalId":8688,"journal":{"name":"Autoimmunity","volume":"58 1","pages":"2445557"},"PeriodicalIF":3.3,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142891604","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
AutoimmunityPub Date : 2025-12-01Epub Date: 2025-02-19DOI: 10.1080/08916934.2025.2461703
Muhammad O Rahman, Andre Sulen, Lars Breivik, Silke Appel, Mark S Anderson, Bergithe E Oftedal, Anette S B Wolff, Eystein S Husebye
{"title":"Anti-perilipin-1 autoantibodies in autoimmune Addison's disease and related endocrine disorders.","authors":"Muhammad O Rahman, Andre Sulen, Lars Breivik, Silke Appel, Mark S Anderson, Bergithe E Oftedal, Anette S B Wolff, Eystein S Husebye","doi":"10.1080/08916934.2025.2461703","DOIUrl":"https://doi.org/10.1080/08916934.2025.2461703","url":null,"abstract":"<p><p>Immune-mediated lipodystrophy syndromes are rare autoimmune disorders characterized by complete or partial destruction of adipocytes in the body. Recently, autoantibodies against perilipin-1 (PLIN1-autoAbs) have been linked to lipodystrophy. Since various perilipins are expressed in the adrenal cortex and ovaries, we asked whether PLIN1-autoAbs were present in patients with adrenal dysfunction and other autoimmune endocrinopathies. Using a sensitive radiobinding immune assay we analyzed anti-PLIN1-autoAbs in 521 patients with endocrinopathies including Sjögren's syndrome. We identified 22 (4.2%) PLIN1-autoAbs positive patients, of whom 15% had autoimmune polyendocrine syndrome type 1 (4/27), 4% autoimmune Addison's disease and/or autoimmune polyendocrine syndrome type 2 (11/274), 8% type 1 diabetes patients (4/53), and 2% Sjögren's syndrome patients (1/50). However, none of them had known lipodystrophy. In conclusion, PLIN1-autoAbs are found in subgroups of autoimmune endocrinopathies and indicate autoimmunity against adipose tissue, but their pathogenic role if any, remains to be defined. Investigating their role in disease progression and their potential as therapeutic targets could pave the way for novel interventions in autoimmune endocrine diseases.</p>","PeriodicalId":8688,"journal":{"name":"Autoimmunity","volume":"58 1","pages":"2461703"},"PeriodicalIF":3.3,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143456811","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
AutoimmunityPub Date : 2025-12-01Epub Date: 2025-02-12DOI: 10.1080/08916934.2025.2465410
Shan Jin, Junbin Yin, Wei Li, Ni Mao
{"title":"Mitochondrial autophagy-related gene signatures associated with myasthenia gravis diagnosis and immunity.","authors":"Shan Jin, Junbin Yin, Wei Li, Ni Mao","doi":"10.1080/08916934.2025.2465410","DOIUrl":"https://doi.org/10.1080/08916934.2025.2465410","url":null,"abstract":"<p><p>Myasthenia gravis (MG) is a common autoimmune disorder that causes skeletal muscle weakness. Most patients presented with skeletal muscle weakness and endurance decline. Mitophagy refers to removing and interpreting aging or damaged mitochondria in cells. This plays a vital role in maintaining cell homeostasis and normal function. This study explores the role of mitophagy-related genes (GM) in MG. Specifically, we collected the transcriptome data of MG and its control group from the GEO database (Gene Expression Omnibus database). The differentially expressed genes (DEGs) were identified by differential analysis and intersected with GM. Multiple machine learning algorithms were applied to screen and verify the diagnostic genes of intersection genes. In addition, we constructed diagnostic models and nomogram models based on diagnostic genes. The immune landscape of MG was explored by ssGSEA analysis. The correlation between the abundance of immune cell infiltration and diagnostic genes was explored by immune infiltration analysis. Finally, the diagnostic genes were further validated by qPCR experiments.</p>","PeriodicalId":8688,"journal":{"name":"Autoimmunity","volume":"58 1","pages":"2465410"},"PeriodicalIF":3.3,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143405603","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Identification and validation of susceptibility modules and hub genes in polyarticular juvenile idiopathic arthritis using WGCNA and machine learning.","authors":"Junfeng Liu, Jianhui Fan, Hongxiang Duan, Guoming Chen, Weihua Zhang, Pingxi Wang","doi":"10.1080/08916934.2024.2437239","DOIUrl":"https://doi.org/10.1080/08916934.2024.2437239","url":null,"abstract":"<p><strong>Background: </strong>Juvenile idiopathic arthritis (JIA), superseding juvenile rheumatoid arthritis (JRA), is a chronic autoimmune disease affecting children and characterized by various types of childhood arthritis. JIA manifests clinically with joint inflammation, swelling, pain, and limited mobility, potentially leading to long-term joint damage if untreated. This study aimed to identify genes associated with the progression and prognosis of JIA polyarticular to enhance clinical diagnosis and treatment.</p><p><strong>Methods: </strong>We analyzed the gene expression omnibus (GEO) dataset GSE1402 to screen for differentially expressed genes (DEGs) in peripheral blood single nucleated cells (PBMCs) of JIA polyarticular patients. Weighted gene co-expression network analysis (WGCNA) was applied to identify key gene modules, and protein-protein interaction networks (PPIs) were constructed to select hub genes. The random forest model was employed for biomarker gene screening. Functional enrichment analysis was conducted using David's online database, gene ontology (GO), and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis to annotate and identify potential JIA pathways. Hub genes were validated using the receiver operating characteristic (ROC) curve.</p><p><strong>Results: </strong>PHLDA1, EGR3, CXCL2, and PF4V1 were identified as significantly associated with the progression and prognosis of JIA polyarticular phenotype, demonstrating high diagnostic and prognostic assessment value.</p><p><strong>Conclusion: </strong>These genes can be utilized as potential molecular biomarkers, offering valuable insights for the early diagnosis and personalized treatment of JIA polyarticular patients.</p>","PeriodicalId":8688,"journal":{"name":"Autoimmunity","volume":"58 1","pages":"2437239"},"PeriodicalIF":3.3,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142852405","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"N6-methyladenosine modification of THBS1 induced by affluent WTAP promotes Graves' ophthalmopathy progression through glycolysis to affect Th17/Treg balance.","authors":"Lin-Na Li, Jie-Man Wu, Zong-Ji Zheng, Shu-Xian Li, Meng-Yi Cai, Meng-Chen Zou","doi":"10.1080/08916934.2024.2433628","DOIUrl":"https://doi.org/10.1080/08916934.2024.2433628","url":null,"abstract":"<p><p>Graves' ophthalmopathy (GO) obvious manifestation is the imbalance of Th17/Treg. N6-methyladenosine (m6A) methylation is an important regulator of Th17/Treg balance. However, few reports narrate how m6A regulators mediate the role of genes in GO progression. We explored the m6A modification of THBS1 mediated by WTAP, and the mechanism by which THBS1 regulated glycolysis and Th17/Treg balance. A total of 12 peripheral blood (4 GO samples, 4 GH samples, and 4 health samples) were collected to measure the percentage of Th17/Treg in monocytes by flow cytometry. RNA sequencing (RNA-seq) combined with MeRIP sequencing (MeRIP-seq) was used to screen differentially expressed and methylated genes. MeRIP-qPCR was performed to evaluate the m6A abundance of THBS1 after WTAP silencing. Glycolysis of CD4<sup>+</sup> T cells was reflected by the lactate content and glucose uptake. The number of Th17 cells was increased in GO peripheral blood, whereas the Treg cells decreased. RNA-seq acquired 679 differentially expressed genes (308 up-regulated, and 371 down-regulated) in the CD4<sup>+</sup> T cells of GO compared to healthy control. MeRIP-seq identified 3277 m6A peaks between the GO group and the healthy control group, corresponding with 2744 genes (1143 hypermethylated and 1601 hypomethylated). Combined analysis of RNA-seq and MeRIP-seq showed 81 hypermethylated and up-regulated genes. Among the six candidate genes in the PI3K-signaling pathway, THBS1 was the most significantly differentially expressed and hypermethylated. THBS1 silencing resulted in decreased lactate content and glucose uptake in CD4<sup>+</sup> T cells. WTAP was significantly upregulated in CD4<sup>+</sup> T cells of GO, and WTAP silencing significantly reduced m6A abundance and expression of THBS1. Upregulated and hypermethylated THBS1 mediated by WTAP promoted glycolysis of CD4<sup>+</sup> T cells, affected Th17/Treg balance, and facilitated GO progression. We provided a novel potential target for GO treatment and revealed the molecular mechanism of WTAP and THBS1 in GO under the m6A perspective.</p>","PeriodicalId":8688,"journal":{"name":"Autoimmunity","volume":"58 1","pages":"2433628"},"PeriodicalIF":3.3,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142845663","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
AutoimmunityPub Date : 2025-12-01Epub Date: 2024-12-23DOI: 10.1080/08916934.2024.2441992
Jing Jing Wang, Ming Wei Lin, Dan Suan, Dimitra Beroukas, Tom P Gordon, Adrian Y S Lee
{"title":"Clinical correlations of serum anti-dsDNA immunoglobulin subfamilies in patients with systemic lupus erythematosus (SLE).","authors":"Jing Jing Wang, Ming Wei Lin, Dan Suan, Dimitra Beroukas, Tom P Gordon, Adrian Y S Lee","doi":"10.1080/08916934.2024.2441992","DOIUrl":"https://doi.org/10.1080/08916934.2024.2441992","url":null,"abstract":"<p><p>Systemic lupus erythematosus (SLE) is an extremely heterogenous autoimmune disorder. A key biomarker, the double stranded (ds) DNA autoantibody, provides diagnostic specificity for SLE. We analyzed anti-dsDNA by mass spectrometry (MS) to determine if ascertaining the autoantibody's heavy chain variable region (IGHV) may hold any clinical relevance. A cross-sectional study of 32 SLE patients (75% female) in a single center was performed. Serum anti-dsDNA was subjected to MS analyses. Obtained IGHV subfamilies were correlated with active clinical features of SLE, as determined by medical record reviews. We established significant associations with the presence of IGHV3-15 and active neuropsychiatric lupus (relative risk [RR] 5.71); IGHV3-21, IGHV3-23 and IGHV4-34 and leukopenia (RR 13.70, 2.14 and 10.29 respectively); and IGHV3-23 and serositis (RR 2.41) and cutaneous lesions (RR 2.82). This study provides the first evidence for the clinical benefits of deep anti-dsDNA profiling through MS, and provides an avenue for improving predictive medicine for SLE patients. Future studies with a greater number of patients, and to determine if these subfamilies have direct pathogenic properties are required.</p>","PeriodicalId":8688,"journal":{"name":"Autoimmunity","volume":"58 1","pages":"2441992"},"PeriodicalIF":3.3,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142880919","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Genome-wide analysis of abnormal splicing regulators and alternative splicing involved in immune regulation in systemic lupus erythematosus.","authors":"Bing Xu, Yuan Liu, Guangfeng Chen, Ping Jiang, Yuan Qu, Mengjie Wang, Xiliang Kao","doi":"10.1080/08916934.2024.2448463","DOIUrl":"https://doi.org/10.1080/08916934.2024.2448463","url":null,"abstract":"<p><p>Systemic lupus erythematosus (SLE) is an autoimmune disease with complex clinical manifestations and no current cure. Alternative splicing (AS) plays a key role in SLE by regulating immune-related genes, but its genome-wide regulatory mechanisms remain unclear. To investigate the involvement of abnormal splicing regulators and AS events in the immune regulation of SLE. Transcriptome data from the SLE dataset GSE162828 were analyzed for differential gene expression and AS events using bioinformatics tools. Immune infiltration analysis was conducted with CIBERSORT, and co-expression of key splicing factors (SFs) and AS events was assessed using SUVA software. A total of 5144 differentially expressed genes and 73 SFs were identified. Significant immune cell differences were observed between SLE and controls, highlighting SFs such as HNRNPDL, RBM47, TIA1, SSB, and DHX15. Eighty-three AS events were identified, with IRF9 and PTPRC emerging as key regulatory events linked to SLE. Dysregulated SFs influence AS in immune-related genes, affecting immune cell composition and SLE progression. These findings offer potential new therapeutic targets for modulating the immune microenvironment in SLE.</p>","PeriodicalId":8688,"journal":{"name":"Autoimmunity","volume":"58 1","pages":"2448463"},"PeriodicalIF":3.3,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142913262","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
AutoimmunityPub Date : 2025-12-01Epub Date: 2025-02-05DOI: 10.1080/08916934.2025.2461072
Laurence Morel
{"title":"Animal models of autoimmunity: a relentless pursuit of accurate pre-clinical models.","authors":"Laurence Morel","doi":"10.1080/08916934.2025.2461072","DOIUrl":"https://doi.org/10.1080/08916934.2025.2461072","url":null,"abstract":"","PeriodicalId":8688,"journal":{"name":"Autoimmunity","volume":"58 1","pages":"2461072"},"PeriodicalIF":3.3,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143254482","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
AutoimmunityPub Date : 2025-12-01Epub Date: 2024-12-18DOI: 10.1080/08916934.2024.2441134
Kangqi Ji, Lingfei Wang, Weijie Liu, Genfeng Li, Xiaoyu Lian, Jun Fan, Chen Song, Yanpeng Jian
{"title":"IDO1-mediated M2 macrophage polarization alleviates the progression of ankylosing spondylitis.","authors":"Kangqi Ji, Lingfei Wang, Weijie Liu, Genfeng Li, Xiaoyu Lian, Jun Fan, Chen Song, Yanpeng Jian","doi":"10.1080/08916934.2024.2441134","DOIUrl":"https://doi.org/10.1080/08916934.2024.2441134","url":null,"abstract":"<p><p>Indoleamine 2,3-dioxygenase 1 (IDO1) plays an anti-inflammatory role in autoimmune disease. However, its specific function in ankylosing spondylitis (AS) remain unclear. This study aimed to investigate the potential role of IDO1 in AS. Immunofluorescence, RT-qPCR, and western blot assays were employed to measure gene expression, while ELISA was used to quantify the release of M1 macrophage and M2 macrophage markers. CCK-8, EdU, flow cytometry, ALP staining, and Alizarin red staining (ARS) assays were conducted for functional analysis. JASPAR predicted the binding sites between PPARγ and the promoter, which were further validated by luciferase and ChIP assays. Our findings revealed that the expression of IDO1 was markedly elevated in AS patients. IDO1 overexpression promoted the proliferation of THP-1 cells and M2 macrophage polarization. Conversely, IDO1 knockdown facilitated the osteogenic differentiation of BMSCs. Furthermore, IDO1-mediated upregulation of PPARγ modulated RUNX2 transcription. PPARγ overexpression counteracted the effects of IDO1 knockdown, thereby inhibiting the osteogenic differentiation of BMSCs. In conclusion, the IDO1/PPARγ/RUNX2 signaling pathway may protect against AS by promoting M2 macrophage polarization and inhibiting osteogenic differentiation.</p>","PeriodicalId":8688,"journal":{"name":"Autoimmunity","volume":"58 1","pages":"2441134"},"PeriodicalIF":3.3,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142845662","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}