{"title":"Cgas deficiency promotes tumor growth by supporting B cell persistence and angiogenesis.","authors":"Papasara Chantawichitwong, Sarinya Kumpunya, Tossapon Wongtangprasert, Peerapat Visitchanakun, Trairak Pisitkun, Prapaporn Pisitkun","doi":"10.1016/j.cellimm.2025.105036","DOIUrl":"https://doi.org/10.1016/j.cellimm.2025.105036","url":null,"abstract":"<p><p>The cGAS sensor activates the STING/IFN signaling pathway, which is crucial for antiviral and antitumor responses. This study aims to investigate the cGAS-mediated immune responses in tumorigenesis using the MC-38 tumor model. MC38-tumor models were established in wild-type (WT) and Cgas-deficient mice to investigate immunophenotypes and cellular mechanisms involved in tumor progression. Cgas<sup>-/-</sup> mice exhibited significantly larger tumors and reduced survival compared to WT mice. Tumors in Cgas<sup>-/-</sup> mice showed increased fibrosis and neovascularity. WT mice mounted a more robust T-cell-mediated antitumor response, with higher levels of NK and effector T cells, while Cgas<sup>-/-</sup> mice showed an expansion of B cells, including regulatory B cells producing IL-10. B cells from tumor-bearing Cgas<sup>-/-</sup> mice demonstrated enhanced survival in the tumor-conditioned medium than those from WT mice. B cell depletion significantly reduced tumor size in WT mice but had minimal effect in Cgas<sup>-/-</sup> mice, where fibrosis and tumor vasculature persisted. Notably, despite B cell depletion, B cells remained in the tumors of Cgas<sup>-/-</sup> mice, in contrast to WT mice, where depletion correlated with increased CD8<sup>+</sup> T cell infiltration. Upregulation of Tgfb1, Tlr7, Tlr9, and Tnfrsf13c in tumors of Cgas<sup>-/-</sup> mice suggested a tumor microenvironment (TME) that promotes B cell survival. Furthermore, Cgas<sup>-/-</sup> B cells promoted angiogenesis, as indicated by enhanced endothelial tube formation. cGAS deficiency fosters tumor growth by reducing the antitumor response, promoting a pro-tumor microenvironment, and supporting B cell survival. The Cgas<sup>-/-</sup> B cells enhance angiogenesis and are resistant to B cell depletion, contributing to tumor progression.</p>","PeriodicalId":9795,"journal":{"name":"Cellular immunology","volume":"418 ","pages":"105036"},"PeriodicalIF":2.9,"publicationDate":"2025-09-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145198502","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":"EGCG remodels the TGF-β cervical cancer micro-environment towards immune responsiveness","authors":"Jayapradha Gnanagurusamy , Rajalakshmi Sabanayagam , Sneha Krishnamoorthy , Vidya Balasubramanian , Sridhar Muthusami","doi":"10.1016/j.cellimm.2025.105027","DOIUrl":"10.1016/j.cellimm.2025.105027","url":null,"abstract":"<div><h3>Background</h3><div>Exploring the role of immune modulators alongside TGF-β in cervical cancer (CC) and PBMCs may improve the understanding of targeted treatment strategies.</div></div><div><h3>Methods</h3><div>We analyzed expression, overall survival (OS), correlation and tumor infiltration of PD-L1, CD55 and CD46, as well as immune cell fractions in CC patients using OncoDB, TIMER 2.0 and TCIA. RT-PCR and western blotting was performed to assess PD-L1, CD55 and CD46 expression. Viability, mitored, apoptosis and MMP-2 were evaluated in CC cells co-cultured with PBMCs. Morphology, crystal violet staining, ROS and MMP-2 were examined in SiHa spheroids.</div></div><div><h3>Results</h3><div>PD-L1 was upregulated, CD55 was significantly increased and CD46 showed no significant difference in HPV16 positive compared with HPV negative individuals. Elevated PD-L1, CD55 and CD46 were associated with reduced OS in HPV16 positive individuals. PD-L1 and CD55 showed moderate positive and negative correlation with TGF-β, respectively, whereas CD46 correlation was negligible. Immune fractions including M1 macrophages (31 %), M2 macrophages (17 %), CD8 T cells (21 %), NK cells (10 %), were linked to reduced OS. TGF-β, PD-L1 and CD46 infiltration were positively corelated with CD8+ T cells. In CC cells, TGF-β stimulation increased PD-L1, while decreasing CD55 and CD46, reducing viability, metabolic activity and inducing apoptosis in HPV (+) co-cultures. EGCG treatment under TGF-β, reduced PD-L1, CD55 and CD46 expression, decreased viability, metabolic activity and MMP-2 secretion, while inducing apoptosis in SiHa co-culture. In 3D spheroids, EGCG inhibited proliferation and MMP-2 activity while increasing ROS production.</div></div><div><h3>Conclusion</h3><div>EGCG, by targeting TGF-β and modulating PD-L1 and mCRPs, represents a promising candidate for immunotherapeutic development in CC.</div></div>","PeriodicalId":9795,"journal":{"name":"Cellular immunology","volume":"418 ","pages":"Article 105027"},"PeriodicalIF":2.9,"publicationDate":"2025-09-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145128348","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}
Jia Yi , Sangmu Quji , Luxuan Guo , Zhongqiu Chai , Xianbin Kong , Jingyan Meng
{"title":"Exploring novel strategies of oncolytic viruses and gut microbiota to enhance CAR-T cell therapy for colorectal cancer","authors":"Jia Yi , Sangmu Quji , Luxuan Guo , Zhongqiu Chai , Xianbin Kong , Jingyan Meng","doi":"10.1016/j.cellimm.2025.105026","DOIUrl":"10.1016/j.cellimm.2025.105026","url":null,"abstract":"<div><div>Colorectal cancer (CRC), ranking as the third most prevalent malignant tumor globally (accounting for 10.0 % of new cancer cases) and the second leading cause of cancer-related deaths (9.4 % of cancer mortality), continues to escalate in incidence, posing a significant threat to human health. Although conventional therapies such as surgery, radiotherapy, and chemotherapy remain the clinical mainstay, their efficacy in improving patient survival and quality of life has reached a plateau, necessitating the exploration of novel therapeutic approaches. Chimeric antigen receptor (CAR) T-cell therapy has emerged as a highly promising approach for cancer treatment. Notably, the complexity of the solid tumor microenvironment (TME) presents challenges for the application of CAR-T therapy in CRC, including antigen heterogeneity, immune suppression, and off-target toxicity. However, the development of multi-target CAR-T cells and their combination with immunomodulatory drugs holds significant clinical potential. Furthermore, in recent years, oncolytic virus (OV) therapy has garnered substantial attention due to its unique antitumor mechanisms. Our study demonstrates that OVs can precisely target CRC tissues, inducing tumor cell apoptosis through selective infection and intracellular replication while concurrently activating systemic antitumor immune responses and inhibiting angiogenesis, thereby achieving multidimensional therapeutic effects. Further investigations reveal that OVs can serve as gene delivery vectors for therapeutic molecules or synergize with chimeric antigen receptor T-cell (CAR-T) therapy and immune checkpoint inhibitors to significantly enhance treatment efficacy. Simultaneously, gut microbiota, a critical regulator of CRC progression, can influence both CAR-T and OVs therapies through metabolic modulation and immune remodeling. Building upon these mechanisms, this review innovatively proposes a tripartite “OVs-gut microbiota-CAR-T\" strategy: OVs may reprogram the immunosuppressive TME and release tumor antigens to enhance CAR-T infiltration and activity, while concurrent modulation of gut microbiota could further alleviate immunosuppression and reduce treatment toxicity, establishing a bidirectional synergistic loop. This interdisciplinary integration strategy may provide a groundbreaking approach to overcome current therapeutic limitations in CRC and advance precision tumor immunotherapy to new frontiers.</div></div>","PeriodicalId":9795,"journal":{"name":"Cellular immunology","volume":"417 ","pages":"Article 105026"},"PeriodicalIF":2.9,"publicationDate":"2025-09-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145091219","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":"Follicular helper T cells and fibrotic diseases","authors":"Yixuan Ma , Yue Zhou , Miao Yu","doi":"10.1016/j.cellimm.2025.105024","DOIUrl":"10.1016/j.cellimm.2025.105024","url":null,"abstract":"<div><div>T follicular helper (Tfh) cells constitute a functionally specialized subset of CD4+ T lymphocytes that orchestrate germinal center (GC) responses, critically regulating B cell destination and the development of long-term humoral immunity. Emerging evidence implicates Tfh cells as key mediators in the pathogenesis of multiple fibrotic disorders across multiple organ systems, including systemic sclerosis (SSc), hepatic fibrosis, chronic kidney disease, idiopathic pulmonary fibrosis (IPF) and IgG4-Related Disease (IgG4-RD). This review synthesizes current understanding of Tfh cell biology in fibrosis, aiming to provide deeper insights into their involvement in pathogenesis and identify potential new therapeutic targets.</div></div>","PeriodicalId":9795,"journal":{"name":"Cellular immunology","volume":"418 ","pages":"Article 105024"},"PeriodicalIF":2.9,"publicationDate":"2025-09-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145157017","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}
Mohua Liu, Xiao Wang, Xiaoya Qu, Yao Wang, Xihui Shen, Lei Xu
{"title":"Divergent metabolic rewiring shapes altered innate immunity","authors":"Mohua Liu, Xiao Wang, Xiaoya Qu, Yao Wang, Xihui Shen, Lei Xu","doi":"10.1016/j.cellimm.2025.105025","DOIUrl":"10.1016/j.cellimm.2025.105025","url":null,"abstract":"<div><div>Both trained immunity (TRIM) and endotoxin tolerance (ET) initiate similar metabolic reprogramming characterized by enhanced glycolysis following an initial stimulus. However, TRIM exhibited heightened immune activation upon restimulation, whereas ET showed suppressed innate immune response. This divergence is attributed to distinct metabolic intermediates accumulated after the initial stimulation. In TRIM, metabolites like fumarate and glutamine derivatives accumulate, reinforcing pro-inflammatory epigenetic modifications. Conversely, ET is characterized by increased itaconate and lactate levels, promoting anti-inflammatory epigenetic changes and metabolic paralysis. This review highlights metabolic intermediates as key regulators of innate immune fate decisions, presenting avenues for targeted immune modulation.</div></div>","PeriodicalId":9795,"journal":{"name":"Cellular immunology","volume":"417 ","pages":"Article 105025"},"PeriodicalIF":2.9,"publicationDate":"2025-09-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145019900","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}
Xin Wang , Xu Sun , Xinmeng Yang , Guixia Xu , Yuan Wang , Mingming Jin , Chao Sun
{"title":"Exosomes from hypoxic pretreated BMSCs attenuate primary Sjögren's syndrome-induced skin injury via PPARγ-mediated M2-like macrophage activation","authors":"Xin Wang , Xu Sun , Xinmeng Yang , Guixia Xu , Yuan Wang , Mingming Jin , Chao Sun","doi":"10.1016/j.cellimm.2025.105023","DOIUrl":"10.1016/j.cellimm.2025.105023","url":null,"abstract":"<div><h3>Background</h3><div>Sjögren's syndrome (SS) is an autoimmune disorder identified by a triad of sicca symptoms, pain, and fatigue. SS-induced skin injury seriously affects people's health but remains unsolved. Accumulating investigations have confirmed that exosomes (Exos) originating from bone marrow mesenchymal stem cells (BMSCs) can bolster the stressed microenvironment and tissue repair. Present study aimed to unravel therapeutic effects regarding BMSC Exos on SS-induced skin injury.</div></div><div><h3>Methods</h3><div>In this study, an SS mouse model was constructed, and exosomes from BMSCs (Exos) and hypoxic pretreated BMSCs (HExos) were isolated. The therapeutic effects of exosomes in SS were identified using ELISA, immunohistochemistry, and immunofluorescence. High-throughput sequencing (HTS) was utilized to characterize differentially expressed genes between Exos and HExos.</div></div><div><h3>Results</h3><div>The data showed that Exos, especially HExo treatments, affected the inhibition of SS-induced inflammatory factor expression, cell apoptosis, ROS deposition, and collagen loss. HTS and RT-qPCR detection showed PPARγ functioned importantly for HExo-mediated protective effects against SS-induced skin injury. The in vitro experiment using RAW confirmed that PPARγ expression inhibited LPS-induced M1-like macrophage activation, which was confirmed using the PPARγ antagonist T0070907. PPARγ upregulation improved therapeutic effects regarding Exos upon skin injury in SS mice by promoting M2-like macrophage activation.</div></div><div><h3>Conclusion</h3><div>Taken together, our study found that exosomes from hypoxic pretreated BMSCs attenuated primary Sjögren's syndrome-induced skin injury via PPARγ delivery and promoted M2-like macrophage activation.</div></div>","PeriodicalId":9795,"journal":{"name":"Cellular immunology","volume":"418 ","pages":"Article 105023"},"PeriodicalIF":2.9,"publicationDate":"2025-09-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145157462","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":"METTL3 positively regulates the development and cytotoxicity of human embryonic stem cells-derived NK cells.","authors":"Xiaofeng Yin, Zhaohui Zhang, Jiaxing Qiu, Yuxing Gong, Qinghua Bi, Meng Meng, Qiangqiang Lai, Hongchen Wang, Shaochang Zhou, Yuan Gao, Lingling Zhang, Wei Wu, Liang Song, Junping Wang, Fangjie Wang, Zhaoyang Zhong, Youcai Deng","doi":"10.1016/j.cellimm.2025.105011","DOIUrl":"10.1016/j.cellimm.2025.105011","url":null,"abstract":"<p><p>Human embryonic stem cell-derived NK (hESC-NK) cells or induced pluripotent stem cell derived NK cells have demonstrated efficacy and safety in clinical trials for cancer therapy and serve as a valuable tool for studying the mechanisms of human NK cell development and effector functions. We previously demonstrated that the methylase METTL3 was essential for the development and effector functions of murine NK cells, but its role in human NK cells remained unknown. Herein, we constructed an H1 ESC strain with reduced METTL3 expression using lentivirus-delivered short hairpin (sh) RNA and generated hESC-NK cells via a two-stage differentiation system. Our findings demonstrated that METTL3 knockdown in hESCs reduced the proportion of hematopoietic stem and progenitor cells (HSPCs, CD34<sup>+</sup> cells) during embryoid bodies (EBs) formation, and impaired subsequent differentiation into mature NK cells. Moreover, ESC-NK cells derived from shMETTL3-ESC (called shMETTL3-ESC-NK) showed impaired anti-tumor activity, evidenced by downregulation of mRNA and protein levels of critical effectors (perforin, granzyme B and IFN-γ) and reduced cytotoxicity against target cells. Furthermore, both mRNA and protein levels of T-BET and EOMES were significantly down-regulated in shMETTL3-ESC-NK cells. These transcription factors are critical for NK cell development and cytotoxicity, and their downregulation may underlie the maturation defects of shMETTL3-ESC-NK cells. Collectively, our study elucidates that METTL3 promotes the development, maturation and cytotoxicity of hESC-NK cells, recapitulating previous reports in murine NK cells.</p>","PeriodicalId":9795,"journal":{"name":"Cellular immunology","volume":"415-416 ","pages":"105011"},"PeriodicalIF":2.9,"publicationDate":"2025-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144803699","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}
Hao-Chien Hung , Tsui-Lien Mao , Ke-Hung Tsui , Ming-Huei Fan , Ainani Priza Minhalina , Chao-Lien Liu
{"title":"A synergy of CD133 overexpression and TGF-β supplementation in tumorigenesis of ovarian cancer cell lines in a three-dimensional sphere forming model","authors":"Hao-Chien Hung , Tsui-Lien Mao , Ke-Hung Tsui , Ming-Huei Fan , Ainani Priza Minhalina , Chao-Lien Liu","doi":"10.1016/j.cellimm.2025.105015","DOIUrl":"10.1016/j.cellimm.2025.105015","url":null,"abstract":"<div><h3>Background</h3><div>Ovarian cancer (OC) is a highly lethal gynecological malignancy, mainly due to chemoresistance and tumor recurrence. Cancer stem cells (CSCs) may be responsible for chemoresistance, and CSC has become a new target for treatment. In this study, we aimed to develop a three-dimensional (3D) OC model with well-recapitulated stemness in the tumor microenvironment (TME).</div></div><div><h3>Results</h3><div>We observed that the niche-like environment associated with CSC properties is characterized by the presence of CD133-positive cells during OC sphere induction. The cancer-associated fibroblast (CAF)-integrated 3D multicellular OC model recapitulates enhanced tumorigenicity and cytokine-mediated invasiveness more than the 2D monolayer culture. Chemoresistance of the 3D OC model is also acquired. In addition, the in vivo growth of an established xenograft model with a 3D CAF-integrated OC sphere exhibits proper stemness features and full cancer-associated markers for tumorigenesis.</div><div>After transduction of the CD133 gene into OC cells, gene ontology (GO) and KEGG pathway enrichment analyses reveal that cytokine-mediated endothelial mesenchymal transition (EMT) is possibly responsible for chemotherapy resistance and tumor progression, and enhanced PAR1, CXCR4, and PD-L1 expressions are also observed. In addition, we found that engineered chimeric antigen receptor (CAR)-T cells targeting PAR1 demonstrated significant in vitro cytotoxicity toward chemoresistant OC sphere with CD133 overexpression.</div></div><div><h3>Conclusions</h3><div>Taken together, our results show that a CD133-3D OC sphere recaptures TME that mimics a real late-stage OC condition, and it can act as a useful platform with mechanism-verifying in vitro and in vivo experiments in researching OC chemotherapy, immunotherapy, and cell therapy to discover new therapeutic approaches.</div></div>","PeriodicalId":9795,"journal":{"name":"Cellular immunology","volume":"417 ","pages":"Article 105015"},"PeriodicalIF":2.9,"publicationDate":"2025-08-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144919967","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":"Establishment of a multi-parameter flow cytometry method to identify and characterize neutrophil extracellular traps","authors":"Weixiang Liu , Jieming Ping , Ning Wu","doi":"10.1016/j.cellimm.2025.105014","DOIUrl":"10.1016/j.cellimm.2025.105014","url":null,"abstract":"<div><div>Neutrophil extracellular traps (NETs) are web-like decondensed DNA filaments released by activated neutrophils, decorated with antimicrobial proteins such as myeloperoxidase (MPO) and elastase. Although several methods exist to evaluate NETs formation, including fluorescent microscopy or scanning electron microscopy (SEM), and flow cytometry, each has inherent limitations that restrict widespread application. Given the increasing relevance of NETs in various pathophysiological contexts, we sought to develop a simple, specific, objective and cost-effective flow cytometry-based method to assess NETs both <em>in vitro</em> and <em>in vivo</em>. Our approach leverages multi-parametric flow cytometry to simultaneously evaluate cell size, granularity, DNA decondensation, histone citrullination, and intracellular MPO. This method enables reliable detection of NETs in purified neutrophils as well as in tissue samples. Its performance was validated in parallel with conventional microscopy, confirming specificity and reproducibility. Notably, this FACS-based method is faster, more economical, and free from observer-bias, making it especially well-suited for both research and clinical sample analysis.</div></div>","PeriodicalId":9795,"journal":{"name":"Cellular immunology","volume":"417 ","pages":"Article 105014"},"PeriodicalIF":2.9,"publicationDate":"2025-08-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144895389","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}
Xuebi Wang , Wei Wang , Yabin Liu , Peng Wang , Qin Huang , Di Tian , Yixin Liao , Zhigang Yi , Feng Li , Yuanjia Tang , Chen Zhao , Xiaohong Fan , Yun Ling
{"title":"Establishing a clinical prediction model of neurosyphilis via a lumbar-puncture-free nomogram","authors":"Xuebi Wang , Wei Wang , Yabin Liu , Peng Wang , Qin Huang , Di Tian , Yixin Liao , Zhigang Yi , Feng Li , Yuanjia Tang , Chen Zhao , Xiaohong Fan , Yun Ling","doi":"10.1016/j.cellimm.2025.105012","DOIUrl":"10.1016/j.cellimm.2025.105012","url":null,"abstract":"<div><h3>Objectives</h3><div>Neurosyphilis (NS) can cause a range of central nervous system (CNS) damage, from asymptomatic states to severe mental disorders. While lumbar puncture is a reliable diagnostic method for NS, it is often poorly accepted due to its invasive nature, particularly by patients with mild symptoms. This study aims to develop a prediction model for the early diagnosis of NS that without requiring lumbar puncture.</div></div><div><h3>Methods</h3><div>Clinical data, including imaging, routine blood tests, immune markers, HIV status and cerebrospinal fluid (CSF) examination, were collected from the Shanghai Public Health Clinical Center from 2021 to 2023,. The dataset was randomly split into training and validation sets in an 8:2 ratio. Univariate analysis, the least absolute shrinkage and selection operator (LASSO) and multivariate logistic regression were used to identify significant predictors.</div></div><div><h3>Results</h3><div>Out of 1078 suspected patients, 702 confirmed syphilis patients with positive <em>Treponema pallidum</em> specific antibody (anti-TP) and positive <em>Treponema Pallidum</em> Particle Agglutination (TRUST) in serum were selected. Among them, 246 patients were diagnosed as NS based on the positive anti-TP in CSF, while 456 patients were classified as non-neurosyphilis (NNS). In the NS group, cerebral ischemia/infarction, ataxia, decreased vision, mental/behavioral disorders, memory impairment, high levels of anti-TP and TRUST (>1:16), elevated red blood cell (RBC) and platelet (PLT) counts, and shorter prothrombin time (PT) were identified. A nomogram was established based on independent prognostic factors. The receiver operating characteristic (ROC) curves and calibration curves showed high predictive accuracy, and clinical decision curve analysis (DCA) indicated good clinical applicability.</div></div><div><h3>Conclusions</h3><div>The developed nomogram offers a reliable and non-invasive method for early identification of NS in syphilis patients, regardless of their HIV or immune status.</div></div>","PeriodicalId":9795,"journal":{"name":"Cellular immunology","volume":"415 ","pages":"Article 105012"},"PeriodicalIF":2.9,"publicationDate":"2025-08-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144865995","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}