1
|
Zhang M, Ma J, Edwards R, Li M. The dynamics of CD4+ T cell proliferation and regulation. JOURNAL OF BIOLOGICAL DYNAMICS 2025; 19:2458867. [PMID: 39881560 DOI: 10.1080/17513758.2025.2458867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 01/21/2025] [Indexed: 01/31/2025]
Abstract
We use mathematical modeling to study the proliferation dynamics of CD4+ T cells within an immune response. This proliferation is driven by the autocrine reaction of helper T cells and interleukin-2 (IL-2), and regulated by natural regulatory T cells (nTregs). Previous studies suggested that a fratricidal mechanism is necessary to eliminate helper T cells post-infection. Contrary to this, our mathematical analysis establishes that the depletion of these cells is due to two pivotal factors: the saturation in the proliferation rate of helper CD4+ T cells at high IL-2 concentrations, and the activation rate of nTregs outpacing their death rate. This yields an excitable process, such that the proliferation starts once the helper T cell population passes a threshold. Additionally, we find that when the proliferation of nTregs lags behind their mortality, induced regulatory T cells (iTregs) are crucial to curbing the proliferation of helper CD4+ T cells.
Collapse
Affiliation(s)
- Mingran Zhang
- College of Information Science and Technology, Donghua University, Shanghai, People's Republic of China
| | - Junling Ma
- Department of Mathematics and Statistics, University of Victoria, Victoria, BC, Canada
| | - Roderick Edwards
- Department of Mathematics and Statistics, University of Victoria, Victoria, BC, Canada
| | - Meili Li
- School of Mathematics and Statistics, Donghua University, Shanghai, People's Republic of China
| |
Collapse
|
2
|
Yu Z, Tang X, Chen Z, Hu Y, Zhang S, Guo C, Gu J, Shi Y, Gong Y. Role of ADAM10/17-Mediated Cleavage of LAG3 in the Impairment of Immunosuppression in Psoriasis. J Invest Dermatol 2025; 145:1385-1395.e8. [PMID: 39571889 DOI: 10.1016/j.jid.2024.10.606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 10/02/2024] [Accepted: 10/04/2024] [Indexed: 12/25/2024]
Abstract
Despite extensive research on immune activation regulatory mechanisms, studies on immune suppression in psoriasis are limited. LAG3, a newly identified immune checkpoint, plays a crucial role in modulating immune responses and maintaining T-regulatory cell function. However, its involvement in psoriasis is unclear. We show that psoriasis is associated with reduced LAG3 expression in CD4 T cells and T-regulatory cells. Further analysis revealed that the decline in LAG3 levels was linked to ADAM10/17-mediated proteolytic cleavage, which was upregulated in psoriasis. Clinical utilization of the IL-17A antagonist secukinumab, along with the in vivo and in vitro IL-17A-induced models, supported the potential of IL-17A to induce ADAM10/17 expression and trigger LAG3 cleavage. Through the Jurkat cell model, IL-17A was found to regulate ADAM10/17 expression by activating FOXM1. In addition, treatment with the ADAM10/17 inhibitor GW280264X showed ameliorative effects on psoriasis-like mouse models and lipopolysaccharide-induced inflammation. Collectively, the findings of this study uncover the immune regulatory role of the ADAM10/17-LAG3 axis in psoriasis and highlight the therapeutic potential of targeting ADAM10/17 for psoriasis treatment.
Collapse
Affiliation(s)
- Zengyang Yu
- Department of Dermatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China; Institute of Psoriasis, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Xinyi Tang
- Department of Dermatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China; Institute of Psoriasis, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Zeyu Chen
- Institute of Psoriasis, Tongji University School of Medicine, Shanghai, People's Republic of China; Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Yifan Hu
- Institute of Psoriasis, Tongji University School of Medicine, Shanghai, People's Republic of China; Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Shuqin Zhang
- Department of Dermatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China; Institute of Psoriasis, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Chunyuan Guo
- Institute of Psoriasis, Tongji University School of Medicine, Shanghai, People's Republic of China; Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Jun Gu
- Department of Dermatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China; Institute of Psoriasis, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Yuling Shi
- Institute of Psoriasis, Tongji University School of Medicine, Shanghai, People's Republic of China; Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China.
| | - Yu Gong
- Department of Dermatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China; Institute of Psoriasis, Tongji University School of Medicine, Shanghai, People's Republic of China.
| |
Collapse
|
3
|
Kibet M, Abebayehu D. Crosstalk between T cells and fibroblasts in biomaterial-mediated fibrosis. Matrix Biol Plus 2025; 26:100172. [PMID: 40226302 PMCID: PMC11986236 DOI: 10.1016/j.mbplus.2025.100172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 02/28/2025] [Accepted: 03/19/2025] [Indexed: 04/15/2025] Open
Abstract
Biomaterial implants are a critical aspect of our medical therapies and biomedical research and come in various forms: stents, implantable glucose sensors, orthopedic implants, silicone implants, drug delivery systems, and tissue engineered scaffolds. Their implantation triggers a series of biological responses that often times lead to the foreign body response and subsequent fibrotic encapsulation, a dense ECM-rich capsule that isolates the biomaterial and renders it ineffective. These responses lead to the failure of biomaterials and is a major hurdle to overcome and in promoting their success. Much attention has been given to macrophage populations for the inflammatory component of these responses to biomaterials but recent work has identified an important role of T cells and their ability to modulate fibroblast activity and vice versa. In this review, we focus on T cell-fibroblast crosstalk by exploring T cell subsets, critical signaling pathways, and fibroblast populations that have been shown to dictate biomaterial-mediated fibrosis. We then highlight emerging technologies and model systems that enable new insights and avenues to T cell-fibroblast crosstalk that will improve biomaterial outcomes.
Collapse
Affiliation(s)
- Mathew Kibet
- Department of Biomedical Engineering, School of Engineering and Medicine, University of Virginia, Charlottesville, VA 22908, United States
| | - Daniel Abebayehu
- Department of Biomedical Engineering, School of Engineering and Medicine, University of Virginia, Charlottesville, VA 22908, United States
| |
Collapse
|
4
|
Yang Y, Xu J, Lu Y, Tang Z, He J. Association of Immune Cell Phenotypes With Oral Cancer: A Two-Sample Mendelian Randomisation Study. Int Dent J 2025; 75:1808-1817. [PMID: 40239604 PMCID: PMC12022481 DOI: 10.1016/j.identj.2025.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/24/2025] [Accepted: 03/12/2025] [Indexed: 04/18/2025] Open
Abstract
OBJECTIVES The purpose of this study is to assess the potential causal relationship between immune cell phenotype and oral cancer using Mendelian randomisation analysis. METHODS A two-sample Mendelian randomisation (MR) analysis using summary statistics from genome-wide association studies in European populations was conducted to explore causal relationships between immune cell phenotypes and the risk of oral cancer. Inverse-variance weighting, MR-EGGER, simple mode, weighted median, and weighted mode were applied for MR analysis. Sensitivity analyses, including the Steiger test, Cochran's Q test, Egger intercept, and leave-one-out analysis, were performed to assess the robustness of the results. Additionally, colocalisation analysis was carried out to further validate causal associations. RESULTS A total of 21 immune cell phenotypes were identified as risk factors for oral cancer, while 6 immune cell phenotypes demonstrated protective effects. Sensitivity analyses indicated a lack of robustness in four causal relationships. Genetic variants at rs9469077 on chr6 might be shared between CD28-CD127-CD25++CD8br AC of regulatory T cells and oral cancer. CONCLUSION This MR study provides evidence for a strong association between immune cells and oral cancer, highlighting specific immune cell phenotypes as significant risk factors for the development of oral cancer. These findings offer a foundation for future precision immunotherapy strategies for oral cancer. Further studies are required to confirm the relationship between immune cells and oral cancer risk and to elucidate the underlying mechanisms. CLINICAL RELEVANCE This study confirms the potential relationship between specific immune cell phenotypes and oral cancer, providing theoretical support for future immunotherapy against oral cancer.
Collapse
Affiliation(s)
- Yanran Yang
- Department of Stomatology, Chengdu Seventh People's Hospital (Affiliated Cancer Hospital of Chengdu Medical College), Chengdu, China.
| | - Jiamin Xu
- Department of Stomatology, Chengdu Seventh People's Hospital (Affiliated Cancer Hospital of Chengdu Medical College), Chengdu, China
| | - Yanzhu Lu
- Department of Stomatology, Chengdu Seventh People's Hospital (Affiliated Cancer Hospital of Chengdu Medical College), Chengdu, China
| | - Zhenxing Tang
- Department of Stomatology, Chengdu Seventh People's Hospital (Affiliated Cancer Hospital of Chengdu Medical College), Chengdu, China
| | - Jiajun He
- Department of Stomatology, Chengdu Seventh People's Hospital (Affiliated Cancer Hospital of Chengdu Medical College), Chengdu, China
| |
Collapse
|
5
|
Wang YX, Ma YD, Li HH, Duo WJ, Jin QW, Zhou KJ, Gao YR, He JN, Xie YJ, Chu L, Yang XD. Schistosoma japonicum cystatin attenuated CLP-induced sepsis in mice though inducing tolerogenic dendritic cells and regulatory T cells. Comp Immunol Microbiol Infect Dis 2025; 120:102345. [PMID: 40344985 DOI: 10.1016/j.cimid.2025.102345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/12/2025] [Accepted: 04/22/2025] [Indexed: 05/11/2025]
Abstract
Sepsis is a life-threatening complication caused by the overwhelming immune response to bacterial infection leading to the fatal organ damage and even death. Helminth infections modulate host's immune system through secreting functional proteins to reduce host immune attack as a survival strategy, therefore have been used for the therapy of some inflammatory or autoimmune diseases. Sj-Cys is a cysteine protease inhibitor secreted by Schistosoma japonicum exerting strong immunomodulatory function which has been used to treat sepsis, however, the mechanism underlying the therapeutic efficacy has not been fully elucidated. In this study, we expressed Sj-Cys as recombinant protein (rSj-Cys) in prokaryotic system and rSj-Cys was used to incubate with mouse bone marrow derived dendritic cells (BMDCs) in vitro. Our study revealed that rSj-Cys was able to induce differentiation of BMDCs to tolerant property (TolDCs). Adoptive transfer of rSj-Cys induced-TolDCs into mice with cecal ligation and puncture (CLP)-induced sepsis conferred a significant therapeutic effect on CLP-induced sepsis in mice with reduced mortality and vital organ damage. The therapeutic effect of Sj-Cys-induced TolDCs was associated with upregulation of CD3+CD4+CD25+Foxp3+ regulatory T cells (Tregs) and reduced inflammatory cytokines IL-6 and TNF-α and boosted level of regulatory cytokines IL-10 and TGF-β. The results identified in this study further suggest rSj-Cys has the potential to be developed into a drug substance for the treatment of inflammatory or autoimmune diseases due to its immunomodulatory effect on tolerant dendritic cells and regulatory T cells.
Collapse
Affiliation(s)
- Yi-Xiang Wang
- Basic Medical College of Bengbu Medical University, Bengbu 233000, China; Anhui Key Laboratory of Infection and Immunity of Bengbu Medical University, Bengbu 233000, China.
| | - Yi-Dan Ma
- Basic Medical College of Bengbu Medical University, Bengbu 233000, China; Anhui Key Laboratory of Infection and Immunity of Bengbu Medical University, Bengbu 233000, China.
| | - Hui-Hui Li
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical University, Bengbu 233000, China.
| | - Wen-Juan Duo
- Basic Medical College of Bengbu Medical University, Bengbu 233000, China; Anhui Key Laboratory of Infection and Immunity of Bengbu Medical University, Bengbu 233000, China; Nan Jing Zi Jin Hospital, Nan Jin 210007, China.
| | - Qi-Wang Jin
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical University, Bengbu 233000, China.
| | - Kai-Jun Zhou
- Basic Medical College of Bengbu Medical University, Bengbu 233000, China.
| | - Yan-Ran Gao
- Basic Medical College of Bengbu Medical University, Bengbu 233000, China.
| | - Jun-Nan He
- Basic Medical College of Bengbu Medical University, Bengbu 233000, China.
| | - Yu-Jie Xie
- Basic Medical College of Bengbu Medical University, Bengbu 233000, China.
| | - Liang Chu
- Anhui Key Laboratory of Infection and Immunity of Bengbu Medical University, Bengbu 233000, China; Second Affiliated Hospital of Bengbu Medical University, Bengbu 233000, China.
| | - Xiao-Di Yang
- Basic Medical College of Bengbu Medical University, Bengbu 233000, China; Anhui Key Laboratory of Infection and Immunity of Bengbu Medical University, Bengbu 233000, China.
| |
Collapse
|
6
|
Phillip West A, McGuire PJ. Tipping the balance: innate and adaptive immunity in mitochondrial disease. Curr Opin Immunol 2025; 95:102566. [PMID: 40424975 DOI: 10.1016/j.coi.2025.102566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 05/01/2025] [Accepted: 05/05/2025] [Indexed: 05/29/2025]
Abstract
Mitochondrial diseases (MtD) provide a unique window into the complex interplay between metabolism and immune function. These rare disorders, caused by defects in oxidative phosphorylation, result in bioenergetic deficiencies that disrupt multiple organ systems. While traditionally studied for their metabolic impact, MtD also profoundly affect the immune system, altering both innate and adaptive responses. This review explores how mitochondrial dysfunction shapes immune dysregulation, influencing thymocyte maturation, regulatory T cells, and B cell function while also driving innate immune activation through mitochondrial DNA instability and type I interferon signaling. Additionally, MtD highlight an emerging overlap between inborn errors of metabolism and inborn errors of immunity, revealing shared pathways that connect mitochondrial dysfunction to immune deficiencies and inflammatory disease. Studying MtD not only advances our understanding of immunometabolism but also provides critical insights into more common inflammatory and autoimmune conditions, offering potential therapeutic targets that extend beyond rare mitochondrial disorders.
Collapse
Affiliation(s)
| | - Peter J McGuire
- National Human Genome Research Institute, Bethesda, MD 20892, USA.
| |
Collapse
|
7
|
Li T, Liu S, Wang S, Sun S, Ji F, Li M, Zhang Y. Identification of metabolic reprogramming-related key genes in hepatocellular carcinoma after transcatheter arterial chemoembolization treatment. Discov Oncol 2025; 16:861. [PMID: 40404896 PMCID: PMC12098233 DOI: 10.1007/s12672-025-02606-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Accepted: 05/06/2025] [Indexed: 05/24/2025] Open
Abstract
BACKGROUND Metabolic reprogramming plays an important role in therapeutic efficacy of hepatocellular carcinoma (HCC). However, the metabolic reprogramming-related key genes associated with transcatheter arterial chemoembolization (TACE) treatment sensitivity in HCC remain further investigation. METHODS We analyzed data from public databases, The Cancer Genome Atlas and Gene Expression Omnibus, as well as metabolism-related genes (MRGs), to identify key genes associated with TACE treatment sensitivity. Further analysis was conducted on the relationship between key genes and immune cell infiltration, HCC-related genes, regulatory network construction, nomogram construction, and drug sensitivity analysis. Finally, the expression of key genes was validated based on databases and in vitro RT-qPCR. RESULTS Four key genes (CDC20, LPCAT1, PON1, and SPP1) associated with TACE treatment sensitivity were identified. Increased CDC20, LPCAT1, and SPP1 and reduced PON1 were found in tumor tissues than normal tissues, as well as in advanced patients than early-stage patients. Lower expression of CDC20, LPCAT1, and SPP1, and higher expression of PON1 were detected in responsive patients than non-responsive patients. Patients with high expression of CDC20, LPCAT1, and SPP1, and low expression of PON1 had poor prognosis. They were also correlated with tumor immune microenvironment and sensitivity to multiple chemotherapy drugs. The expressions of key genes at the gene and protein levels were validated. CONCLUSIONS Our study provided systematic insights into identification of biomarkers for TACE treatment sensitivity in HCC.
Collapse
Affiliation(s)
- Tongfei Li
- Department of Interventional Radiology, The Second Affiliated Hospital of Shandong First Medical University, Tai'an, 271000, China
| | - Shujuan Liu
- Department of Oncology, Jinan Seventh People's Hospital, Jinan, 250132, China
| | - Shengjun Wang
- Department of Oncology, Jinan Seventh People's Hospital, Jinan, 250132, China
| | - Shan Sun
- Department of Oncology, Jinan Seventh People's Hospital, Jinan, 250132, China
| | - Feng Ji
- Department of Oncology, Shanxian Dongda Hospital, Heze, 274399, China
| | - Mingliang Li
- Automation Department, Jigang Group International Engineering Technology Co., Ltd, Jinan, 250098, China
| | - Yong Zhang
- School of Preventive Medicine, Shandong First Medical University, No. 6699 Qingdao Road, Jinan, 250117, Shandong, People's Republic of China.
| |
Collapse
|
8
|
Yang J, Zhang H, Wang W, Yin Q, He X, Tao D, Wang H, Liu W, Wang Y, Dong Z, Chen X, Li B. CD80 Antibody and MTX Co-Engineered Extracellular Vesicles Targets CD80 + Macrophages to Suppress Inflammation and Alleviate Chronic Inflammatory Diseases. Int J Nanomedicine 2025; 20:6379-6398. [PMID: 40416732 PMCID: PMC12103861 DOI: 10.2147/ijn.s517357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 05/13/2025] [Indexed: 05/27/2025] Open
Abstract
Introduction Aberrant interaction between innate immune and adaptive immune cells can disrupt tissue homeostasis, consequently triggering chronic inflammatory diseases such as rheumatoid arthritis (RA) and periodontitis (PD). Pro-inflammatory macrophages serve as critical mediators in the early immune response, constituting a major population of CD80+ cells, while anti-inflammatory macrophages modulating inflammatory processes through the secretion of transforming growth factor-beta (TGF-β). This cytokine facilitates the differentiation of peripheral regulatory T cells (Tregs) and contributes to the establishment of immune tolerance. However, there are no definitive therapies to reshape the tissue homeostasis between innate immune and adaptive immune cells. Methods (1) anti-CD80-MTX-EVs was obtained by gradient centrifugation, which were characterized by TEM and DLS, and the associated membrane proteins were identified by Western Blot. (2) The mouse bone marrow-derived macrophages were co-cultured separately with EVs, anti-CD80-EVs, and anti-CD80-MTX-EVs in vitro, and the expression of CD80 on the macrophages surface as well as the proportion of Treg cell generation were detected. (3) EVs, anti-CD80-EVs and anti-CD80-MTX-EVs were injected into mice models of arthritis and periodontitis for treatment, the therapeutic effect was evaluated by the expressions of related cytokines, staining of HE, the proportion of CD80+ macrophages and the phenotypic differentiation of T cells in the tissues. Results We successfully constructed engineered EVs (anti-CD80-MTX-EVs) targeting inflammatory macrophages for intracellular MTX delivering, which inducing the anti-inflammatory transformation while upregulating the expression of TGF-β of macrophages. Furthermore, our findings demonstrate that anti-CD80-MTX-EVs effectively reduce CD80+ macrophage levels, promote Treg cell generation, and inhibit Th1 cell production in vivo. Conclusion In this study, the anti-CD80-MTX-Evs demonstrated significant therapeutic effects in both rheumatoid arthritis and periodontitis models through a triple mechanism: reducing CD80+ macrophage population, enhancing Treg cell differentiation, and suppressing Th1 cell development. Overall, this study presents an innovative strategy for resolving inflammation within chronic inflammatory diseases.
Collapse
Affiliation(s)
- Jianhua Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi’an, People’s Republic of China
| | - Handan Zhang
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi’an Jiaotong University, Xi’an, 710049, People’s Republic of China
| | - Wenzhe Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi’an, People’s Republic of China
| | - Qiqi Yin
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi’an Jiaotong University, Xi’an, 710049, People’s Republic of China
| | - Xiaoning He
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi’an, People’s Republic of China
| | - Dihao Tao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi’an, People’s Republic of China
| | - Hanzhe Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi’an, People’s Republic of China
| | - Wenhao Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi’an, People’s Republic of China
| | - Yiming Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi’an, People’s Republic of China
| | - Zhiwei Dong
- Department of Oral and Maxillofacial Surgery, College of Stomatology, Xi’an Jiaotong University, Xi’an, People’s Republic of China
| | - Xin Chen
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi’an Jiaotong University, Xi’an, 710049, People’s Republic of China
| | - Bei Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi’an, People’s Republic of China
| |
Collapse
|
9
|
Liu M, Deng H, Liu C, Wang L, Liao Z, Li D, Chen Y, Li J, Dong J, Sun X, Wang C, Huang L, Dong L, Xiao J. Islet transplantation in immunomodulatory nanoparticle-remodeled spleens. Sci Transl Med 2025; 17:eadj9615. [PMID: 40397715 DOI: 10.1126/scitranslmed.adj9615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 10/08/2024] [Accepted: 04/29/2025] [Indexed: 05/23/2025]
Abstract
Islet transplantation is a promising therapy for insulin-dependent diabetes. However, immune rejection and insufficient vascularization hinder the survival and function of transplanted islets. Here, we show effective engraftment of vascularized and functional mouse and rat islets transplanted into biomaterial-remodeled spleens of nonimmunosuppressed rodents and human islets transplanted into the remodeled spleens of nonhuman primates (NHPs) on varying degrees of immunosuppression. We found evidence that konjac glucomannan-modified silica nanoparticles (KSiNPs) remodeled the spleen into an extracellular matrix (ECM)-rich, immunosuppressive niche to support the survival of syngeneic or xenogeneic islets. Transplanted islets in the remodeled spleens showed improved engraftment, neovascularization, and functionality and restored normoglycemia in streptozotocin (STZ)-induced type 1 diabetic models in the mice and macaques, with stable insulin and C-peptide secretion in mice for 90 days and macaques for 28 days. KSiNP injection and islet transplantation into macaque spleens under B-ultrasound guidance were preclinically feasible. These findings highlight the safety and effectiveness of spleen tissue remodeling in supporting the survival and function of transplanted islets, providing a promising strategy for treating type 1 diabetes mellitus (T1DM).
Collapse
Affiliation(s)
- Mi Liu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Department of Wound Healing of the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, 999078, Macau SAR, China
- Affiliated Cixi Hospital, Wenzhou Medical University, Cixi, 315300, China
| | - Huiming Deng
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Department of Wound Healing of the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
| | - Chunyan Liu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Lintao Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Zhongkai Liao
- Department of Organ Transplantation, Second Affiliated Hospital of Hainan Medical University, Haikou, 570216, China
| | - Desheng Li
- Department of Organ Transplantation, Second Affiliated Hospital of Hainan Medical University, Haikou, 570216, China
| | - Yan Chen
- Department of Oncology of the First Affiliated Hospital and Cancer Institute, Hainan Medical University, Haikou, 570102, China
| | - Jianhui Li
- Division of Hepatobiliary Pancreatic Surgery, Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jianhui Dong
- Institute of Transplantation Medicine, Second Affiliated Hospital of Guangxi Medical University; Guangxi Clinical Research Center for Organ Transplantation; Guangxi Key Laboratory of Organ Donation and Transplantation, Nanning 530007, China
| | - Xuyong Sun
- Institute of Transplantation Medicine, Second Affiliated Hospital of Guangxi Medical University; Guangxi Clinical Research Center for Organ Transplantation; Guangxi Key Laboratory of Organ Donation and Transplantation, Nanning 530007, China
| | - Chunming Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, 999078, Macau SAR, China
| | - Ling Huang
- School of Hainan Provincial Drug Safety Evaluation Research Center, Hainan Medical University, Haikou, 571199, China
- Center for Pharmacovigilance of Hainan Province, Hainan Medical Products Administration, Haikou, 570216, China
| | - Lei Dong
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Jian Xiao
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), School of Pharmaceutical Science, Department of Wound Healing of the First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
| |
Collapse
|
10
|
Peter A, Vermeulen M, Van Delen M, Dams A, Peeters S, De Reu H, Marei WFA, Berneman ZN, Cools N. Physiological Oxygen Levels in the Microenvironment Program Ex Vivo-Generated Conventional Dendritic Cells Toward a Tolerogenic Phenotype. Cells 2025; 14:736. [PMID: 40422239 DOI: 10.3390/cells14100736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2025] [Revised: 05/13/2025] [Accepted: 05/14/2025] [Indexed: 05/28/2025] Open
Abstract
Dendritic cells (DCs) are critical regulators of immune homeostasis, balancing tolerance and immunity through antigen presentation and T cell modulation. While the influence of hypoxia (<2% O2) on DC function in pathological settings is well-documented, the impact of physiological O2 levels remains underexplored. This study investigates the role of physioxia (4% O2) in programming mature DCs toward a tolerogenic phenotype compared to atmospheric conditions (21% O2) typically present in in vitro assays. DC cultures generated under 4% O2 exhibited a reduced monocyte-to-DC transformation rate, increased lactate production, a semi-mature surface marker profile, and increased surface expression of the tolerance-associated marker ILT4. T cell priming was altered only when atmospheric DCs were co-cultured under physioxia, suggesting an O2-dependent threshold for immunostimulatory capacity. These findings highlight the complexity of O2-dependent mechanisms in DC-T cell interactions, revealing a delicate balance between tolerance and immunogenicity. Our results underscore the need for physiologically relevant O2 conditions in DC research to better reflect in vivo behavior and inform immunotherapy design. Overall, this study advances understanding of how microenvironmental cues shape DC biology, with implications for immune tolerance, autoimmunity, and cancer immunotherapy.
Collapse
Affiliation(s)
- Antonia Peter
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium
| | - Morgane Vermeulen
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium
| | - Mats Van Delen
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium
- Health Department, Flemish Institute for Technological Research (VITO), 2400 Mol, Belgium
| | - Amber Dams
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium
| | - Stefanie Peeters
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium
- Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, 2650 Edegem, Belgium
| | - Hans De Reu
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium
- Flow Cytometry and Sorting Core Facility (FACSUA), University of Antwerp, 2610 Antwerp, Belgium
| | - Waleed F A Marei
- Gamete Research Centre, Laboratory of Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, 2610 Antwerp, Belgium
- Department of Theriogenology, Faculty of Veterinary Medicine, Cairo University, Giza 3725005, Egypt
| | - Zwi N Berneman
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium
- Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, 2650 Edegem, Belgium
| | - Nathalie Cools
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium
- Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, 2650 Edegem, Belgium
- Flow Cytometry and Sorting Core Facility (FACSUA), University of Antwerp, 2610 Antwerp, Belgium
| |
Collapse
|
11
|
Ou M, Cao J, Luo R, Zhu B, Miao R, Yu L, Wang X, Li W, Fu Y, Zhang J, Zhang F, Wang Q, Mei L. Drug-loaded microneedle patches containing regulatory T cell-derived exosomes for psoriasis treatment. Acta Biomater 2025; 198:452-466. [PMID: 40210183 DOI: 10.1016/j.actbio.2025.04.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 03/17/2025] [Accepted: 04/07/2025] [Indexed: 04/12/2025]
Abstract
Psoriasis is a chronic inflammatory skin disease characterized by epidermal hyperplasia, skin inflammation, and immune dysregulation. These factors contribute to the persistent progression of the disease. While addressing excessive keratinocyte proliferation or inhibiting inflammation may provide temporary therapeutic relief, unresolved immune dysregulation often exacerbates the condition. Therefore, comprehensive treatments that alleviate skin symptoms and regulate immune tolerance are urgently required. An ideal treatment would target multiple factors, including keratinocyte proliferation, inflammation, and immune tolerance, while minimizing systemic side effects. In this study, we developed a dissolvable hyaluronic acid microneedle patch containing regulatory T cell (Treg) exosomes loaded with dimethyl fumarate (DMF) (rExo@DMF MNs). DMF acts as an inhibitor of keratinocyte proliferation and an anti-inflammatory agent through NF-κB suppression and Nrf2 activation, inhibiting the production of pro-inflammatory cytokines and the activation of inflammatory cells. Delivering DMF via Treg exosomes enhances its retention at the lesion site. This system inhibits keratinocyte proliferation and migration, reduces pro-inflammatory cytokine release, and alleviates epidermal hyperplasia and inflammation in an imiquimod-induced psoriasis mouse model. Additionally, Treg exosomes modulate immune responses to promote tolerance. rExo@DMF MNs demonstrate immunomodulatory effects by inhibiting T helper 17 (Th17) cells and inducing regulatory immune cells such as Tregs and tolerogenic dendritic cells (tDCs) differentiation. rExo@DMF MNs alleviate skin symptoms and regulate immune cells in the skin, spleen, and lymph nodes, demonstrating both local and systemic immunoregulation with promising therapeutic potential for psoriasis. STATEMENT OF SIGNIFICANCE: Novel therapies are urgently needed to alleviate skin symptoms and regulate immunity, as current psoriasis treatments focus on symptom relief while neglecting the underlying immune dysfunction, resulting in limited efficacy. Moreover, systemic immunosuppression often leads to severe side effects. This study introduces a hybrid microneedle system (rExo@DMF MNs) that alleviates psoriasis symptoms and modulates immune responses locally and systemically. In addition, rExo@DMF MNs penetrate hyperkeratotic skin, ensuring targeted rExo@DMF release while minimizing systemic exposure and side effects. All components of the system, including hyaluronic acid (a key component of the skin matrix), regulatory T cell-derived exosomes, and DMF (a clinically validated drug), exhibit biocompatibility. This comprehensive approach addresses multiple pathogenic factors, promising an effective and safe psoriasis treatment.
Collapse
Affiliation(s)
- Meitong Ou
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China
| | - Jiahui Cao
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China
| | - Ran Luo
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China
| | - Baisong Zhu
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China
| | - Rourou Miao
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China
| | - Liu Yu
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China
| | - Xinyi Wang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China
| | - Wen Li
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China
| | - Yiqiu Fu
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China
| | - Jinxie Zhang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China
| | - Fan Zhang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China.
| | - Qiangsong Wang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China.
| | - Lin Mei
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, PR China.
| |
Collapse
|
12
|
Sureka N, Zaheer S. Regulatory T Cells in Tumor Microenvironment: Therapeutic Approaches and Clinical Implications. Cell Biol Int 2025. [PMID: 40365758 DOI: 10.1002/cbin.70031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/19/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025]
Abstract
Regulatory T cells (Tregs), previously referred to as suppressor T cells, represent a distinct subset of CD4+ T cells that are uniquely specialized for immune suppression. They are characterized by the constitutive expression of the transcription factor FoxP3 in their nuclei, along with CD25 (the IL-2 receptor α-chain) and CTLA-4 on their cell surface. Tregs not only restrict natural killer cell-mediated cytotoxicity but also inhibit the proliferation of CD4+ and CD8+ T-cells and suppress interferon-γ secretion by immune cells, ultimately impairing an effective antitumor immune response. Treg cells are widely recognized as a significant barrier to the effectiveness of tumor immunotherapy in clinical settings. Extensive research has consistently shown that Treg cells play a pivotal role in facilitating tumor initiation and progression. Conversely, the depletion of Treg cells has been linked to a marked delay in tumor growth and development.
Collapse
Affiliation(s)
- Niti Sureka
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| |
Collapse
|
13
|
Jiang X, Wang M, Zou R, Fu M, Fan W, Wang Y, Dai C, Swapnil Z, Wang W, Wu H, Xie K, Liu L, Wang Y, Fan Z, Zhao L. Harnessing Kupffer Cell Metabolic Rewiring: Rapamycin-Gliadin Nanoparticle as a Pivotal Strategy for Immune Tolerance in Celiac Disease. ACS NANO 2025; 19:17462-17477. [PMID: 40302617 DOI: 10.1021/acsnano.4c18354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2025]
Abstract
Celiac disease (CeD), triggered by gliadin exposure, necessitates therapeutic strategies that establish an antigen-specific immune tolerance. This study explores the therapeutic efficacy and mechanism of rapamycin-gliadin composite nanoparticles (PLN-GR) for CeD treatment. In vivo analyses demonstrated the efficient uptake of PLN-GR by antigen-presenting cells (APCs), particularly Kupffer cells and splenic dendritic cells (DCs), driving their tolerogenic phenotypic transformation. In a murine CeD model, PLN-GR administration significantly enhanced gluten tolerance and mitigated intestinal inflammation, as indicated by reduced paw edema and improved histopathological parameters. Mechanistically, PLN-GR induced macrophage metabolic reprogramming from glycolysis to oxidative phosphorylation, concomitant with elevated serum itaconate levels. This metabolic shift potentiated interorgan immunoregulatory crosstalk, expanding PD-L1+ tolerogenic splenic DCs while suppressing pathogenic Th1 cell populations. Bone marrow-derived macrophages (BMDMs) from Acod1-/- mice (deficient in itaconate synthesis) failed to induce DC tolerance upon PLN-GR treatment. However, supplementation with the itaconate derivative 4-octyl itaconate (4-OI) restored PD-L1 expression in DC2.4 cells in vitro, revealing that itaconate induces and stabilizes the tolerant DC phenotype. These findings underscore PLN-GR as a novel nanotherapeutic platform for CeD, achieving gliadin-specific tolerance through hepatic-splenic immunometabolic reprogramming and itaconate-dependent PD-L1 regulation, thereby offering a translatable strategy for autoimmune disease management.
Collapse
Affiliation(s)
- Xiaohan Jiang
- Department of Digestive Endoscopy, Jiangsu Province Hospital and the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
- Department of General Surgery, Jiangsu Province Hospital and the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Min Wang
- Department of Digestive Endoscopy, Jiangsu Province Hospital and the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
- Department of General Surgery, Jiangsu Province Hospital and the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Ruihan Zou
- Department of Digestive Endoscopy, Jiangsu Province Hospital and the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
- Department of General Surgery, Jiangsu Province Hospital and the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Min Fu
- Department of Digestive Endoscopy, Jiangsu Province Hospital and the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
- Gastroenterology Department, The Fourth Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Wentao Fan
- Department of Digestive Endoscopy, Jiangsu Province Hospital and the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
- Gastroenterology Department, The Fourth Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Yao Wang
- Department of Digestive Endoscopy, Jiangsu Province Hospital and the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
- Department of General Surgery, Jiangsu Province Hospital and the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Chenguang Dai
- Department of Gastroenterology, First Afilliated Hospital of Soochow University, Soochow 215000, China
| | - Zaman Swapnil
- Department of Digestive Endoscopy, Jiangsu Province Hospital and the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Wanjun Wang
- Department of Digestive Endoscopy, Jiangsu Province Hospital and the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
- Gastroenterology Department, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou 213000, China
| | - Hao Wu
- Department of Digestive Endoscopy, Jiangsu Province Hospital and the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
- Department of General Surgery, Jiangsu Province Hospital and the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Kunxin Xie
- Pancreas Center, Jiangsu Province Hospital and the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Li Liu
- Department of Digestive Endoscopy, Jiangsu Province Hospital and the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
- Department of General Surgery, Jiangsu Province Hospital and the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Yan Wang
- Department of Digestive Endoscopy, Jiangsu Province Hospital and the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
- The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Ili & Jiangsu Joint Institute of Health, Ili 835800, China
| | - Zhining Fan
- Department of Digestive Endoscopy, Jiangsu Province Hospital and the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
- Department of General Surgery, Jiangsu Province Hospital and the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| | - Lili Zhao
- Department of Digestive Endoscopy, Jiangsu Province Hospital and the First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
14
|
Paktinat S, Gravett MG, Tobey C, Kirby A, Horner W, Shaffer R, Fialkow M, Nguyen NP, Gornalusse GG, Kalatehjari M, Hughes SM, Hladik F, Vojtech L. Extracellular vesicles from human semen induce unique tolerogenic phenotypes in vaginal dendritic cells and regulatory T lymphocytes. Front Immunol 2025; 16:1564002. [PMID: 40421022 PMCID: PMC12104210 DOI: 10.3389/fimmu.2025.1564002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 04/17/2025] [Indexed: 05/28/2025] Open
Abstract
Introduction The regulation of immune responses to promote tolerance to the fetus is critical for successful pregnancy. An understudied aspect of this process is the initiation of regulation pre-conception via exposure to semen. Our study aimed to understand how semen impacts recipient dendritic cells (DCs) and their subsequent role in shaping CD4 T cell differentiation. Methods Monocyte-derived DCs (MoDCs) were exposed to semen extracellular vesicles (SEV) or vesicle-depleted semen plasma (VDSP). Phenotypic and functional markers were analyzed using flow cytometry. We also exposed epithelial sheets from vaginal tissue to SEV and VDSP, and measured the number and marker expression of emigrating cells. Finally, we tested how SEV- or VDSP-exposed DCs altered CD4 T cell differentiation by co-culturing exposed MoDCs or tissue emigrated cells with autologous naïve CD4 T cells. Results MoDCs exhibited a significant increase of CD141, CD1a, CD38, and ILT4 expression when exposed to SEV or VDSP. A unique feature of semen-treated MoDCs was expression of indoleamine 2,3-dioxygenase (IDO), a potent contributor to the induction of regulatory T cells (Tregs). SEV but not VDSP significantly increased the emigration of intraepithelial DCs. Additionally, SEV significantly enhanced the expression of multiple immunoregulatory markers in the emigrated DCs. After co-culture, we observed significantly more FOXP3+ Tregs expressing high levels of TIGIT in the groups that were initially exposed to SEV. Discussion These findings indicate that exposure to SEV induces a tolerogenic program in DCs that can direct differentiation of a unique memory Treg subset, primed for expansion and presumably destined to support a successful pregnancy.
Collapse
Affiliation(s)
- Shahrokh Paktinat
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States
| | - Michael G. Gravett
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States
| | - Cara Tobey
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States
| | - Anna Kirby
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States
| | - Whitney Horner
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States
| | - Rebecca Shaffer
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States
| | - Michael Fialkow
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States
| | - Nam Phuong Nguyen
- Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA, United States
| | - Germán G. Gornalusse
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States
| | - Maryam Kalatehjari
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States
| | - Sean M. Hughes
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States
| | - Florian Hladik
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States
- Department of Medicine, University of Washington, Seattle, WA, United States
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Lucia Vojtech
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States
| |
Collapse
|
15
|
Zhang W, Cui N, Su F, Liu M, Li B, Sun Y, Zeng Y, Yang B, Kuang H, Wang Q. Effects of Rehmanniae Radix Praeparata polysaccharides on LPS-induced immune activation in mice based on gut microbiota, metabolomics and transcriptomics. Int J Biol Macromol 2025; 311:143981. [PMID: 40339850 DOI: 10.1016/j.ijbiomac.2025.143981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/27/2025] [Accepted: 05/05/2025] [Indexed: 05/10/2025]
Abstract
This study explored the immunomodulatory effects of Rehmanniae Radix Praeparata polysaccharides (RP) on LPS-induced immune activation. RP, characterized as a heteropolysaccharide (6.34 kDa and 4.63 kDa) rich in galactose and glucose, was administered to LPS-challenged BALB/c mice at 25 mg/kg and 50 mg/kg doses. Results showed RP significantly reduced pro-inflammatory cytokines (TNF-α, IL-6), lowered oxidative stress (MDA), and boosted antioxidant enzymes (SOD, GSH-Px). It restored splenic structure, mitigated apoptosis, and suppressed the TNF-α/NF-κB/IL-6 pathway. Metabolomics linked RP to sphingolipid metabolism, while gut microbiota analysis revealed increased beneficial bacteria and elevated SCFAs. Transcriptomics confirmed RP's immune regulation via TNF signaling. These findings demonstrate RP's potential in alleviating immune overactivation by modulating inflammation, gut microbiota, and SCFA production, suggesting therapeutic promise for immune-related diseases.
Collapse
Affiliation(s)
- Wensen Zhang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, 24 Heping Road, Xiangfang District, Harbin 150040, China
| | - Na Cui
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, 24 Heping Road, Xiangfang District, Harbin 150040, China
| | - Fazhi Su
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, 24 Heping Road, Xiangfang District, Harbin 150040, China
| | - Meng Liu
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, 24 Heping Road, Xiangfang District, Harbin 150040, China
| | - Biao Li
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, 24 Heping Road, Xiangfang District, Harbin 150040, China
| | - Yanping Sun
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, 24 Heping Road, Xiangfang District, Harbin 150040, China
| | - Yuanning Zeng
- Guangdong Engineering Technology Research Center for Standardized Processing of Chinese Materia Medica (Guangdong Pharmaceutical University, School of Chinese Materia Medica), Guangdong 510006, China
| | - Bingyou Yang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, 24 Heping Road, Xiangfang District, Harbin 150040, China
| | - Haixue Kuang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, 24 Heping Road, Xiangfang District, Harbin 150040, China
| | - Qiuhong Wang
- Guangdong Engineering Technology Research Center for Standardized Processing of Chinese Materia Medica (Guangdong Pharmaceutical University, School of Chinese Materia Medica), Guangdong 510006, China
| |
Collapse
|
16
|
Kakh M, Doroudchi M, Talepoor A. Induction of Regulatory T Cells After Virus Infection and Vaccination. Immunology 2025. [PMID: 40329764 DOI: 10.1111/imm.13927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 03/04/2025] [Accepted: 03/17/2025] [Indexed: 05/08/2025] Open
Abstract
Vaccines have been proven to be one of the safest and most effective ways to prevent and combat diseases. However, the main focus has been on the evaluation of the potency of effector mechanisms and the lack of adverse effects of vaccine candidates. Recently, the importance of induced regulatory mechanisms of the immune system after vaccination has come to light. With the increase in our knowledge about these regulatory mechanisms including the regulatory T cells (Tregs), we have come to understand the significance of this arm of the immune system in controlling immunopathology and/or diminishing the effectiveness of vaccines, especially viral vaccines. Tregs play a dual role during infectious diseases by limiting immune-mediated pathology and also contributing to chronic pathogen persistence by decreasing effector immunity and clearance of infection. Tregs may also affect immune responses after vaccination primarily by inhibiting antigen presenting cell function such as cytokine secretion and co-stimulatory molecule expression as well as effector T (Teff) and B cell function. In this article, we review the current knowledge on the induction of Tregs after several life-threatening virus infections and their available vaccines to bring them to the spotlight and emphasise that studying viral-induced antigen-specific Tregs will help us improve the effectiveness and decrease the immunopathology or side effects of viral vaccines. Trial Registration: ClinicalTrials.gov identifier: NCT04357444.
Collapse
Affiliation(s)
- MansourehKarimi Kakh
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehrnoosh Doroudchi
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - AtefeGhamar Talepoor
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Autoimmune Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
17
|
Ssedyabane F, Niyonzima N, Ngonzi J, Najjuma JN, Namuli A, Okeny C, Nuwashaba D, Birungi A, Kajabwangu R, Randall TC, Castro CM, Lee H, Tusubira D. Diagnostic Accuracy of Serum P16ink4A and FOX-P3 Concentrations for Detection of Cervical Lesions Among Women Attending a Cervical Cancer Clinic in Western Uganda: A Case-Control Study. Anal Cell Pathol (Amst) 2025; 2025:1931921. [PMID: 40365511 PMCID: PMC12074841 DOI: 10.1155/ancp/1931921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 04/19/2025] [Indexed: 05/15/2025] Open
Abstract
Introduction: Expression of P16ink4A and FOXP3 is correlated with the grades of cervical lesions. In this study, we determined the diagnostic accuracy of serum P16ink4A and FOXP3 concentrations for detection of cervical intraepithelial neoplasia (CIN) and cervical cancer (CC) in a rural setting in Southwestern Uganda. Material and Methods: CIN and CC cases (93 each before treatment), and 93 controls were identified. Clinical and demographic data were documented before quantifying serum P16ink4A and FOXP3 concentrations using quantitative ELISA kits. Cases were confirmed by cytology and/or histology. We employed descriptive statistics, cross-tabulation, and receiver operating curves (ROC) using statistical software for data science (STATA) 17. p-values <0.05 were considered statistically significant. Results: Serum FOXP3 concentration of 0.0545 ng/mL < showed moderate sensitivity (32.22% and 57.78%) for detection of CIN and CC from healthy controls, respectively. It also showed a moderately high specificity of 68.89% for detection of both CIN and CC from healthy controls (AUC-0.6014 and 0.7679, respectively). Serum P16ink4A concentration of 0.946 ng/mL < showed moderate sensitivities (50.00% and 60.00%) and specificities (56.67% and 55.56%) for the detection of CIN and CC from healthy controls, respectively (AUC-0.6085 and 0.7592, respectively). A combination of elevated serum FOXP3 and P16ink4A showed very low sensitivities of 18.89% in detecting CIN from healthy controls and 33.33% for detecting CC from healthy controls. This combination showed high specificity of 83.33% in detecting both CIN and CC from healthy controls (AUC-0.5992 and 0.7642, respectively). Conclusion: Although serum P16ink4A and FOXP3 concentrations showed moderate accuracy, their combination was more specific than sensitive. This combination has a high potential to be applied for diagnosis rather than screening for cervical lesions, at least in the Ugandan population. Combinations of P16ink4A and FOXP3 with other biomarkers could improve diagnostic accuracies. Additionally, studies could be conducted to assess the performance of these biomarkers in the detection of cervical lesions in specific populations, say Human Immunodeficiency Virus (HIV)-positive and HIV-negative populations.
Collapse
Affiliation(s)
- Frank Ssedyabane
- Department of Medical Laboratory Science, Faculty of Medicine, Mbarara University of Science of Science and Technology, Mbarara, Uganda
| | - Nixon Niyonzima
- Department of Medical Laboratory Science, Faculty of Medicine, Mbarara University of Science of Science and Technology, Mbarara, Uganda
- Research and Training Directorate, Uganda Cancer Institute, Kampala, Uganda
| | - Joseph Ngonzi
- Department of Obstetrics and Gynecology, Mbarara University of Science of Science and Technology, Mbarara, Uganda
| | - Josephine Nambi Najjuma
- Department of Nursing, Mbarara University of Science of Science and Technology, Mbarara, Uganda
| | - Alexcer Namuli
- Department of Obstetrics and Gynecology, Mbarara University of Science of Science and Technology, Mbarara, Uganda
| | - Christopher Okeny
- Department of Medical Laboratory Science, Faculty of Medicine, Mbarara University of Science of Science and Technology, Mbarara, Uganda
| | - Doreen Nuwashaba
- Department of Medical Laboratory Science, Faculty of Medicine, Mbarara University of Science of Science and Technology, Mbarara, Uganda
| | - Abraham Birungi
- Department of Pathology, Mbarara University of Science of Science and Technology, Mbarara, Uganda
| | - Rogers Kajabwangu
- Department of Obstetrics and Gynecology, Mbarara University of Science of Science and Technology, Mbarara, Uganda
| | - Thomas C. Randall
- Department of Global Health and Social Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Cesar M. Castro
- Department of Nursing, Mbarara University of Science of Science and Technology, Mbarara, Uganda
- Centre for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston 02114, Massachusetts, USA
- Cancer Centre, Massachusetts General Hospital, Harvard Medical School, Boston 02114, Massachusetts, USA
| | - Hakho Lee
- Centre for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston 02114, Massachusetts, USA
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston 02114, Massachusetts, USA
| | - Deusdedit Tusubira
- Department of Biochemistry, Mbarara University of Science of Science and Technology, Mbarara, Uganda
| |
Collapse
|
18
|
Kart U, Raimbekova A, Yegorov S, Hortelano G. Immune Modulation with Oral DNA/RNA Nanoparticles. Pharmaceutics 2025; 17:609. [PMID: 40430900 DOI: 10.3390/pharmaceutics17050609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 04/02/2025] [Accepted: 04/07/2025] [Indexed: 05/29/2025] Open
Abstract
The oral delivery of DNA/RNA nanoparticles represents a transformative approach in immunotherapy and vaccine development. These nanoparticles enable targeted immune modulation by delivering genetic material to specific cells in the gut-associated immune system, triggering both mucosal and systemic immune responses. Unlike parenteral administration, the oral route offers a unique immunological environment that supports both tolerance and activation, depending on the formulation design. This review explores the underlying mechanisms of immune modulation by DNA/RNA nanoparticles, their design and delivery strategies, and recent advances in their application. Emphasis is placed on strategies to overcome physiological barriers such as acidic pH, enzymatic degradation, mucus entrapment, and epithelial tight junctions. Special attention is given to the role of gut-associated lymphoid tissue in mediating immune responses and the therapeutic potential of these systems in oral vaccine platforms, food allergies, autoimmune diseases, and chronic inflammation. Despite challenges, recent advances in nanoparticle formulation support the translation of these technologies into clinical applications for both therapeutic immunomodulation and vaccination.
Collapse
Affiliation(s)
- Ulpan Kart
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, 53 Kabanbay Batyr Ave., Astana 010000, Kazakhstan
| | - Aigul Raimbekova
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, 53 Kabanbay Batyr Ave., Astana 010000, Kazakhstan
| | - Sergey Yegorov
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, 53 Kabanbay Batyr Ave., Astana 010000, Kazakhstan
| | - Gonzalo Hortelano
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, 53 Kabanbay Batyr Ave., Astana 010000, Kazakhstan
| |
Collapse
|
19
|
Shichijo T, Yasunaga JI. Stratagems of HTLV-1 for persistent infection and the resultant oncogenesis: Immune evasion and clonal expansion. Leuk Res 2025; 152:107680. [PMID: 40120237 DOI: 10.1016/j.leukres.2025.107680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 02/23/2025] [Accepted: 03/12/2025] [Indexed: 03/25/2025]
Abstract
Adult T-cell leukemia-lymphoma (ATL) is one of the most severe malignant T-cell leukemia/lymphomas induced by human T-cell leukemia virus type I (HTLV-1). HTLV-1 persists in the host through stratagems of proliferating infected cells and evading host immunity. HTLV-1 encodes two viral oncogenes, tax and HTLV-1 bZIP factor (HBZ), which are related with protection from cell death and promotion of cell proliferation. In addition, HBZ and the somatic mutations in host genes, such as C-C chemokine receptor 4 (CCR4) and CIC, convert HTLV-1-infected cells into regulatory T (Treg)-like cells, leading to evasion of host immunity. A recent study demonstrated the key mechanisms for clonal expansion of HTLV-1-infected cells; the activation of the transforming growth factor (TGF)-β signaling pathway by HBZ not only converts HTLV-1-infected cells into a Treg-like cells through Foxp3 expression, but also contributes to the proliferation of HTLV-1-infected cells themselves. Due to the longevity induced by HTLV-1 infection, somatic mutations and epigenetic aberrations are accumulated in infected clones, contributing to the oncogenesis of ATL. Collectively, the long-term survival of infected cells enabled by the HTLV-1's stratagems for persistent infection ultimately leads to ATL oncogenesis via the accumulation of genetic/epigenetic abnormalities.
Collapse
Affiliation(s)
- Takafumi Shichijo
- Department of Hematology, Rheumatology and Infectious Diseases, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan.
| | - Jun-Ichirou Yasunaga
- Department of Hematology, Rheumatology and Infectious Diseases, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan.
| |
Collapse
|
20
|
Surico PL, Barone V, Singh RB, Coassin M, Blanco T, Dohlman TH, Basu S, Chauhan SK, Dana R, Di Zazzo A. Potential applications of mesenchymal stem cells in ocular surface immune-mediated disorders. Surv Ophthalmol 2025; 70:467-479. [PMID: 39097173 DOI: 10.1016/j.survophthal.2024.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 07/29/2024] [Accepted: 07/29/2024] [Indexed: 08/05/2024]
Abstract
We explore the interaction between corneal immunity and mesenchymal stem/stromal cells (MSCs) and their potential in treating corneal and ocular surface disorders. We outline the cornea's immune privilege mechanisms and the immunomodulatory substances involved. In this realm, MSCs are characterized by their immunomodulatory properties and regenerative potential, making them promising for therapeutic application. Therefore, we focus on the role of MSCs in immune-mediated corneal diseases such as dry eye disease, corneal transplantation rejection, limbal stem cell deficiency, and ocular graft-versus-host disease. Preclinical and clinical studies demonstrate MSCs' efficacy in promoting corneal healing and reducing inflammation in these conditions. Overall, we emphasize the potential of MSCs as innovative therapies in ophthalmology, offering promising solutions for managing various ocular surface pathologies.
Collapse
Affiliation(s)
- Pier Luigi Surico
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA; Department of Ophthalmology, Campus Bio-Medico University Hospital, Rome 00128, Italy; Cornea Rare Diseases Center, Fondazione Policlinico Campus Bio-Medico, Rome 00128, Italy
| | - Vincenzo Barone
- Department of Ophthalmology, Campus Bio-Medico University Hospital, Rome 00128, Italy; Cornea Rare Diseases Center, Fondazione Policlinico Campus Bio-Medico, Rome 00128, Italy
| | - Rohan Bir Singh
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Marco Coassin
- Department of Ophthalmology, Campus Bio-Medico University Hospital, Rome 00128, Italy; Cornea Rare Diseases Center, Fondazione Policlinico Campus Bio-Medico, Rome 00128, Italy
| | - Tomas Blanco
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Thomas H Dohlman
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Sayan Basu
- Brien Holden Eye Research Centre (BHERC), L. V. Prasad Eye Institute, Hyderabad, Telangana, India
| | - Sunil K Chauhan
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Reza Dana
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Antonio Di Zazzo
- Department of Ophthalmology, Campus Bio-Medico University Hospital, Rome 00128, Italy; Cornea Rare Diseases Center, Fondazione Policlinico Campus Bio-Medico, Rome 00128, Italy.
| |
Collapse
|
21
|
Zhong H, Zhou S, Yin S, Qiu Y, Liu B, Yu H. Tumor microenvironment as niche constructed by cancer stem cells: Breaking the ecosystem to combat cancer. J Adv Res 2025; 71:279-296. [PMID: 38866179 DOI: 10.1016/j.jare.2024.06.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 05/27/2024] [Accepted: 06/09/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND Cancer stem cells (CSCs) are a distinct subpopulation of cancer cells with the capacity to constantly self-renew and differentiate, and they are the main driver in the progression of cancer resistance and relapse. The tumor microenvironment (TME) constructed by CSCs is the "soil" adapted to tumor growth, helping CSCs evade immune killing, enhance their chemical resistance, and promote cancer progression. AIM OF REVIEW We aim to elaborate the tight connection between CSCs and immunosuppressive components of the TME. We attempt to summarize and provide a therapeutic strategy to eradicate CSCs based on the destruction of the tumor ecological niche. KEY SCIENTIFIC CONCEPTS OF REVIEW This review is focused on three main key concepts. First, we highlight that CSCs recruit and transform normal cells to construct the TME, which further provides ecological niche support for CSCs. Second, we describe the main characteristics of the immunosuppressive components of the TME, targeting strategies and summarize the progress of corresponding drugs in clinical trials. Third, we explore the multilevel insights of the TME to serve as an ecological niche for CSCs.
Collapse
Affiliation(s)
- Hao Zhong
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Shiyue Zhou
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Shuangshuang Yin
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, China
| | - Yuling Qiu
- School of Pharmacy, Tianjin Medical University, Tianjin, China.
| | - Bo Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China.
| | - Haiyang Yu
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, China.
| |
Collapse
|
22
|
Zuo G, Li M, Guo X, Wang L, Yao Y, Huang JA, Liu Z, Lin Y. Fu brick tea supplementation ameliorates non-alcoholic fatty liver disease and associated endotoxemia via maintaining intestinal homeostasis and remodeling hepatic immune microenvironment. Food Res Int 2025; 209:116207. [PMID: 40253128 DOI: 10.1016/j.foodres.2025.116207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/27/2025] [Accepted: 03/11/2025] [Indexed: 04/21/2025]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a prevalent disorder of excessive fat accumulation and inflammation in the liver that currently lacks effective therapeutic interventions. Fu brick tea (FBT) has been shown to ameliorate liver damage and modulate gut microbiota dysbiosis in NAFLD, but the potential mechanisms have not been comprehensively elucidated, especailly whether its hepatoprotective effects are determined to depend on the homeostasis of gut microbiota, intestinal barrier function and hepatic immune microenvironment. In this study, our results further demonstrated that FBT not only alleviated NAFLD symptoms and related endotoxemia in high-fat diet (HFD)-fed rats, but also attenuated intestinal barrier dysfunction and associated inflammation, also confirmed in Caco-2 cell experiment. Meanwhile, FBT intervention significantly relieved HFD-induced gut microbiota dysbiosis, characterized by increased diversity and composition, particularly facilitating beneficial microbes, including short chain fatty acids (SCFAs) and bile acids producers, such as Blautia and Fusicatenibacter, and inhibiting Gram-negative bacteria, such as Prevotella_9 and Phascolarctobacterium. Also, the gut microbiota-dependent hepatoprotective effects of FBT were verified by fecal microbiota transplantation (FMT) experiment. Thus, the beneficial moulation of gut microbiota altered by FBT in levels of SCFAs, bile acids and lipopolysaccharides, intestinal barrier function and TLR4/NF-κB pathway contributed to alleviate liver steatosis and inflammation. Additionally, the hepatoprotective effects of FBT was further demonstrated by suppressing Kupffer cell activation and regulating lipid metabolism using an ex vivo model of liver organoid. Therefore, FBT supplementation can maintain intenstinal homeostasis and remodel hepatic immune microenvironment to prevent NAFLD and associated endotoxemia.
Collapse
Affiliation(s)
- Gaolong Zuo
- Key Laboratory of Tea Science of Ministry of Education and Co-Innovation Centre of Education Ministry for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, PR China; National Research Center of Engineering & Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, PR China
| | - Menghua Li
- Key Laboratory of Tea Science of Ministry of Education and Co-Innovation Centre of Education Ministry for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, PR China; National Research Center of Engineering & Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, PR China
| | - Xiaoli Guo
- Key Laboratory of Tea Science of Ministry of Education and Co-Innovation Centre of Education Ministry for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, PR China; National Research Center of Engineering & Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, PR China
| | - Ling Wang
- Key Laboratory of Tea Science of Ministry of Education and Co-Innovation Centre of Education Ministry for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, PR China; National Research Center of Engineering & Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, PR China
| | - Yanyan Yao
- Key Laboratory of Tea Science of Ministry of Education and Co-Innovation Centre of Education Ministry for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, PR China; National Research Center of Engineering & Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, PR China
| | - Jian-An Huang
- Key Laboratory of Tea Science of Ministry of Education and Co-Innovation Centre of Education Ministry for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, PR China; National Research Center of Engineering & Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, PR China; Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha 410128, PR China.
| | - Zhonghua Liu
- Key Laboratory of Tea Science of Ministry of Education and Co-Innovation Centre of Education Ministry for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, PR China; National Research Center of Engineering & Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, PR China; Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha 410128, PR China.
| | - Yong Lin
- Key Laboratory of Tea Science of Ministry of Education and Co-Innovation Centre of Education Ministry for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, PR China; National Research Center of Engineering & Technology for Utilization of Botanical Functional Ingredients, Hunan Agricultural University, Changsha 410128, PR China; Key Laboratory for Evaluation and Utilization of Gene Resources of Horticultural Crops, Ministry of Agriculture and Rural Affairs of China, Hunan Agricultural University, Changsha 410128, PR China.
| |
Collapse
|
23
|
Soto-Heredero G, Gabandé-Rodríguez E, Carrasco E, Escrig-Larena JI, Gómez de Las Heras MM, Delgado-Pulido S, Francos-Quijorna I, Blanco EM, Fernández-Almeida Á, Abia D, Rodríguez MJ, Fernández-Díaz CM, Álvarez-Flores MB, Ramírez de Molina A, Jung S, Del Sol A, Zorita V, Sánchez-Cabo F, Torroja C, Mittelbrunn M. KLRG1 identifies regulatory T cells with mitochondrial alterations that accumulate with aging. NATURE AGING 2025; 5:799-815. [PMID: 40307497 DOI: 10.1038/s43587-025-00855-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 03/27/2025] [Indexed: 05/02/2025]
Abstract
Recent studies using single-cell RNA sequencing technology have uncovered several subpopulations of CD4+ T cells that accumulate with aging. These age-associated T cells are emerging as relevant players in the onset of inflammaging and tissue senescence. Here, based on information provided by single-cell RNA sequencing data, we present a flow cytometry panel that allows the identification of age-associated T cell subsets in systematic larger analysis in mice. We use this panel to evaluate at the single-cell level mitochondrial and senescence marks in the different age-associated CD4+ T cell subpopulations. Our analysis identifies a subpopulation of regulatory T (Treg) cells that is characterized by the extracellular expression of the co-inhibitory molecule killer cell lectin-like receptor subfamily G member 1 (KLRG1) and accumulates with aging in humans and mice. KLRG1-expressing Treg cells display senescence features such as mitochondrial alterations, increased expression of cell-cycle regulators and genomic DNA damage. Functionally, KLRG1+ Treg cells show a reduced suppressive activity in vivo accompanied by a pro-inflammatory phenotype.
Collapse
Affiliation(s)
- Gonzalo Soto-Heredero
- Departamento de Biología Molecular, Facultad de Ciencias, Centro de Biología Molecular 'Severo Ochoa', Universidad Autónoma de Madrid, Madrid, Spain
- Consejo Superior de Investigaciones Científicas, Centro de Biología Molecular 'Severo Ochoa', Universidad Autónoma de Madrid, Madrid, Spain
| | - Enrique Gabandé-Rodríguez
- Departamento de Biología Molecular, Facultad de Ciencias, Centro de Biología Molecular 'Severo Ochoa', Universidad Autónoma de Madrid, Madrid, Spain
- Consejo Superior de Investigaciones Científicas, Centro de Biología Molecular 'Severo Ochoa', Universidad Autónoma de Madrid, Madrid, Spain
| | - Elisa Carrasco
- Departamento de Biología, Facultad de Ciencias, Centro de Biología Molecular 'Severo Ochoa', Universidad Autónoma de Madrid, Madrid, Spain
| | - José Ignacio Escrig-Larena
- Consejo Superior de Investigaciones Científicas, Centro de Biología Molecular 'Severo Ochoa', Universidad Autónoma de Madrid, Madrid, Spain
| | - Manuel M Gómez de Las Heras
- Departamento de Biología Molecular, Facultad de Ciencias, Centro de Biología Molecular 'Severo Ochoa', Universidad Autónoma de Madrid, Madrid, Spain
| | - Sandra Delgado-Pulido
- Consejo Superior de Investigaciones Científicas, Centro de Biología Molecular 'Severo Ochoa', Universidad Autónoma de Madrid, Madrid, Spain
| | - Isaac Francos-Quijorna
- Departamento de Biología Molecular, Facultad de Ciencias, Centro de Biología Molecular 'Severo Ochoa', Universidad Autónoma de Madrid, Madrid, Spain
- Consejo Superior de Investigaciones Científicas, Centro de Biología Molecular 'Severo Ochoa', Universidad Autónoma de Madrid, Madrid, Spain
| | - Eva M Blanco
- Consejo Superior de Investigaciones Científicas, Centro de Biología Molecular 'Severo Ochoa', Universidad Autónoma de Madrid, Madrid, Spain
| | - Álvaro Fernández-Almeida
- Consejo Superior de Investigaciones Científicas, Centro de Biología Molecular 'Severo Ochoa', Universidad Autónoma de Madrid, Madrid, Spain
| | - David Abia
- Consejo Superior de Investigaciones Científicas, Centro de Biología Molecular 'Severo Ochoa', Universidad Autónoma de Madrid, Madrid, Spain
| | - María Josefa Rodríguez
- Servicio de Microscopía Electrónica, Centro de Biología Molecular 'Severo Ochoa', Universidad Autónoma de Madrid, Madrid, Spain
| | | | | | | | - Sascha Jung
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch sur-Alzette, Luxembourg
- CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Bizkaia Technology Park, Derio, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Antonio Del Sol
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch sur-Alzette, Luxembourg
- CIC bioGUNE-BRTA (Basque Research and Technology Alliance), Bizkaia Technology Park, Derio, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Virginia Zorita
- Centro Nacional de Investigaciones Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain
| | - Fátima Sánchez-Cabo
- Centro Nacional de Investigaciones Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain
| | - Carlos Torroja
- Centro Nacional de Investigaciones Cardiovasculares, Instituto de Salud Carlos III, Madrid, Spain
| | - María Mittelbrunn
- Consejo Superior de Investigaciones Científicas, Centro de Biología Molecular 'Severo Ochoa', Universidad Autónoma de Madrid, Madrid, Spain.
| |
Collapse
|
24
|
Sutanto H, Elisa E, Rachma B, Fetarayani D. Gut Microbiome Modulation in Allergy Treatment: The Role of Fecal Microbiota Transplantation. Am J Med 2025; 138:769-777.e3. [PMID: 39855612 DOI: 10.1016/j.amjmed.2025.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/10/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025]
Abstract
The prevalence of allergic diseases has been rising, paralleling lifestyle changes and environmental exposures that have altered human microbiome composition. This review article examines the intricate relationship between the gut microbiome and allergic diseases, emphasizing the potential of fecal microbiota transplantation as a promising novel treatment approach. It explains how reduced microbial exposure in modern societies contributes to immune dysregulation and the increasing incidence of allergies. The discussion also addresses immune homeostasis and its modulation by the gut microbiome, highlighting the shift from eubiosis to dysbiosis in allergic conditions. Furthermore, this article reviews existing studies and emerging research on the role of fecal microbiota transplantation in restoring microbial balance, providing insights into its mechanisms, efficacy, and safety.
Collapse
Affiliation(s)
- Henry Sutanto
- Internal Medicine Study Program, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia; Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Elisa Elisa
- Internal Medicine Study Program, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia; Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Betty Rachma
- Internal Medicine Study Program, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia; Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia
| | - Deasy Fetarayani
- Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya, Indonesia; Division of Allergy and Clinical Immunology, Department of Internal Medicine, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia.
| |
Collapse
|
25
|
Paganelli A, Didona D, Scala E. Cytokine Networks in Lichen Sclerosus: A Roadmap for Diagnosis and Treatment? Int J Mol Sci 2025; 26:4315. [PMID: 40362551 PMCID: PMC12072692 DOI: 10.3390/ijms26094315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Revised: 04/28/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025] Open
Abstract
Lichen sclerosus (LS) is a chronic inflammatory skin disorder primarily affecting the anogenital region, leading to symptoms such as itching, pain, and sexual dysfunction, all of which significantly impact patients' quality of life. Due to the non-specific nature of its early symptoms, diagnosis is often delayed. This review examines the cytokine networks involved in LS, with a focus on immune activation, the role of T-helper (Th)1 cells, and the interaction between inflammatory mediators and the extracellular matrix, particularly in fibrosis. By providing an updated understanding of LS immunopathogenesis, this review highlights key mediators involved in disease progression and offers insights into personalized treatment strategies that may improve patient outcomes. Additionally, current therapeutic approaches and future directions in LS management are discussed.
Collapse
Affiliation(s)
- Alessia Paganelli
- Dermatology Unit, IDI-IRCCS Istituto Dermopatico dell’Immacolata, 00167 Rome, Italy;
| | - Dario Didona
- Rare Diseases Unit, IDI-IRCCS Istituto Dermopatico dell’Immacolata, 00167 Rome, Italy
| | - Emanuele Scala
- Laboratory of Experimental Immunology, IDI-IRCCS Istituto Dermopatico dell’Immacolata, 00167 Rome, Italy
| |
Collapse
|
26
|
Satooka H, Nakamura Y, Hirata T. ROS-dependent SOCS3 upregulation disrupts regulatory T cell stability during autoimmune disease development. Redox Biol 2025; 82:103590. [PMID: 40090133 PMCID: PMC11957609 DOI: 10.1016/j.redox.2025.103590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 03/02/2025] [Accepted: 03/09/2025] [Indexed: 03/18/2025] Open
Abstract
Autoimmune diseases including rheumatoid arthritis (RA) are often associated with high levels of reactive oxygen species (ROS); however, the ROS targets in autoimmunity are diverse and unclear. Using collagen-induced arthritis (CIA) mice as a model for RA, we report that antioxidants markedly suppress joint inflammation, antibody production, and effector T cell responses. We found that the frequency of CD4+ regulatory T cells (Tregs) was reduced in CIA mice, which was reversed by antioxidant treatment, and SOCS3, known to be associated with Treg instability, was upregulated in Tregs from both RA patients and CIA mice. Mechanistically, SOCS3 upregulation was induced by ROS-dependent PTEN oxidation and the resultant Akt/mTOR/STAT3 activation. We further showed that the source of ROS involved in this pathway is NADPH oxidase 2 (Nox2). Nox2 expression was upregulated in Tregs from CIA mice, and Nox2 transduction induced a decrease in Treg frequency that depended on SOCS3 upregulation. This study thus provides a mechanistic understanding of ROS-induced Treg instability and suggests that ROS-dependent disruption of Treg homeostasis underlies the development and progression of autoimmune diseases.
Collapse
Affiliation(s)
- Hiroki Satooka
- Department of Fundamental Biosciences, Shiga University of Medical Science, Otsu, Shiga, 520-2192, Japan
| | - Yuzuki Nakamura
- Department of Fundamental Biosciences, Shiga University of Medical Science, Otsu, Shiga, 520-2192, Japan
| | - Takako Hirata
- Department of Fundamental Biosciences, Shiga University of Medical Science, Otsu, Shiga, 520-2192, Japan.
| |
Collapse
|
27
|
Maeda E, Okimura H, Tanaka Y, Fujii M, Tarumi Y, Kataoka H, Koshiba A, Hamaguchi M, Fukui M, Mori T, Kitawaki J. Adoptive transfer of regulatory T cells inhibits the progression of endometriosis-like lesions in regulatory T-cell-depleted mice. Hum Reprod 2025; 40:926-937. [PMID: 40180333 DOI: 10.1093/humrep/deaf054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 02/26/2025] [Indexed: 04/05/2025] Open
Abstract
STUDY QUESTION Does the restoration of regulatory T cells (Tregs) suppress the progression of endometriosis? SUMMARY ANSWER Adoptive transfer of Tregs suppresses the progression of endometriosis and reduces the levels of helper T (Th)-cell-related and proinflammatory cytokines in mice. WHAT IS KNOWN ALREADY Endometriosis is a chronic inflammatory gynecological disease, which involves multiple immune components. Activated Treg counts decrease in the endometrioma and endometrium of patients with endometriosis, and depletion of Tregs exacerbates endometriosis in mice. STUDY DESIGN, SIZE, DURATION We evaluated the effects of adoptive transfer of Tregs on the progression of endometriosis in mice. We used Foxp3tm3Ayr/J (Foxp3DTR) mice with temporarily ablated Tregs by injecting diphtheria toxin to develop an endometriosis model, which was generated by ovariectomy, estradiol administration and transplantation of uterine fragments from donor mice. Foxp3DTR mice were randomly divided into Treg adoptive transfer (n = 12) and control (n = 11) groups. Tregs were isolated from lymph nodes and spleens of wild-type (WT) mice and were adoptively transferred into mice that were temporarily Treg-depleted. Control mice were injected with vehicle. Treg adoptive transfer was performed on the day of uterine implantation, and a second adoptive transfer was performed after 14 days. Mice were euthanized 28 days after uterine implantation, and blood, peritoneal fluid, spleen, and endometriosis-like lesion samples were collected. PARTICIPANTS/MATERIALS, SETTING, METHODS Foxp3DTR mice were intravenously injected with Tregs isolated from WT mice. The number, total weight, and total volume of the endometriosis-like lesions were evaluated on Day 28 following implantation of uterine fragments. The proportion of Tregs in endometriosis-like lesions, ascites, and peripheral blood was analyzed by flow cytometry. Inflammation in lesions and serum was examined using real-time PCR and ELISA. MAIN RESULTS AND THE ROLE OF CHANCE Injection of Tregs increased their total count and decreased the number (P < 0.0001), weight (P = 0.0021), and volume (P = 0.0010) of endometriosis-like lesions in Foxp3DTR Treg-depleted mice. Furthermore, injection of Tregs decreased the mRNA expression of Th 1-, 2-, and 17-related cytokines, including interferon gamma (P = 0.0101), interleukin (IL)-4 (P = 0.0051), and IL-17 (P = 0.0177), as well as the levels of the proinflammatory cytokine IL-6 (P = 0.0002), in endometriosis-like lesions of Foxp3DTR Treg-depleted mice. LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION Treg-related immune mechanisms in mice may not precisely reflect those in humans. WIDER IMPLICATIONS OF THE FINDINGS Restoration of Tregs may be a useful therapeutic strategy for inhibiting the progression of endometriosis in cases where the decrease in the Treg population is an exacerbating factor. STUDY FUNDING/COMPETING INTEREST(S) This study was partially supported by the Grants-in-Aid for Scientific Research (grant numbers 18K16808 and 20K22983) from the Ministry of Education, Culture, Sports, Science, and Technology, Japan. The sponsor had no role in the study design, collection, analysis and interpretation of data, writing of the report, and decision to submit the article for publication. The authors have no conflicts of interest to disclose.
Collapse
Affiliation(s)
- Eiko Maeda
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hiroyuki Okimura
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yukiko Tanaka
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Maya Fujii
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yosuke Tarumi
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hisashi Kataoka
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Akemi Koshiba
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Masahide Hamaguchi
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Michiaki Fukui
- Department of Endocrinology and Metabolism, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Taisuke Mori
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Jo Kitawaki
- Department of Obstetrics and Gynecology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
28
|
Parolini C. Pathophysiology of bone remodelling cycle: Role of immune system and lipids. Biochem Pharmacol 2025; 235:116844. [PMID: 40044049 DOI: 10.1016/j.bcp.2025.116844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/31/2025] [Accepted: 02/28/2025] [Indexed: 03/15/2025]
Abstract
Osteoporosis is the most common skeletal disease worldwide, characterized by low bone mineral density, resulting in weaker bones, and an increased risk of fragility fractures. The maintenance of bone mass relies on the precise balance between bone synthesis and resorption. The close relationship between the immune and skeletal systems, called "osteoimmunology", was coined to identify these overlapping "scientific worlds", and its function resides in the evaluation of the mutual effects of the skeletal and immune systems at the molecular and cellular levels, in both physiological and pathological states. Lipids play an essential role in skeletal metabolism and bone health. Indeed, bone marrow and its skeletal components demand a dramatic amount of daily energy to control hematopoietic turnover, acquire and maintain bone mass, and actively being involved in whole-body metabolism. Statins, the main therapeutic agents in lowering plasma cholesterol levels, are able to promote osteoblastogenesis and inhibit osteoclastogenesis. This review is meant to provide an updated overview of the pathophysiology of bone remodelling cycle, focusing on the interplay between bone, immune system and lipids. Novel therapeutic strategies for the management of osteoporosis are also discussed.
Collapse
Affiliation(s)
- Cinzia Parolini
- Department of Pharmacological and Biomolecular Sciences, 'Rodolfo Paoletti', via Balzaretti 9 - Università degli Studi di Milano 20133 Milano, Italy.
| |
Collapse
|
29
|
Lin M, Zhang Q, Cong Y. Research Progress of Oral Immune Tolerance Mechanism Induced by Whey Protein. Nutrients 2025; 17:1517. [PMID: 40362825 PMCID: PMC12073718 DOI: 10.3390/nu17091517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 04/27/2025] [Accepted: 04/28/2025] [Indexed: 05/15/2025] Open
Abstract
Cow milk allergy (CMA) is prevalently observed among infants and young children, exerting adverse effects on their growth and quality of life. Oral immune tolerance (OIT) is a more effective method for the prevention and treatment of CMA. The site of OIT is mainly in the gastrointestinal tract, so this article reviews the composition and structural characteristics of intestinal immune system, the molecular mechanisms of immune tolerance by regulatory T cells (Treg), dendritic cells, and gut microbiota. In addition, this paper summarizes the research progress of T cell epitope peptides of β-lactoglobulin and α-lactalbumin in whey protein hydrolysates. The mechanism of OIT induced by whey protein hydrolysate or whey protein combined with other anti-allergic components (phenolic compounds, probiotics, etc.) is overviewed to provide new ideas for the development of hypoallergenic infant formula.
Collapse
Affiliation(s)
| | | | - Yanjun Cong
- College of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| |
Collapse
|
30
|
Paton H, Sarkar P, Gurung P. An overview of host immune responses against Leishmania spp. infections. Hum Mol Genet 2025:ddaf043. [PMID: 40287829 DOI: 10.1093/hmg/ddaf043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 03/18/2025] [Accepted: 03/24/2025] [Indexed: 04/29/2025] Open
Abstract
Leishmania spp. infections pose a significant global health challenge, affecting approximately 1 billion people across more than 88 endemic countries. This unicellular, obligate intracellular parasite causes a spectrum of diseases, ranging from localized cutaneous lesions to systemic visceral infections. Despite advancements in modern medicine and increased understanding of the parasite's etiology and associated diseases, treatment options remain limited to pentavalent antimonials, liposomal amphotericin B, and miltefosine. A deeper understanding of the interactions between immune and non-immune cells involved in the clearance of Leishmania spp. infections could uncover novel therapeutic strategies for this debilitating disease. This review highlights recent progress in elucidating how various cell types contribute to the regulation and resolution of Leishmania spp. infections.
Collapse
Affiliation(s)
- Hanna Paton
- Inflammation Program, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
- Department of Internal Medicine, University of Iowa, 431 Newton Road, Iowa City, IA 52442, United States
- Immunology Graduate Program, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
| | - Prabuddha Sarkar
- Inflammation Program, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
- Department of Internal Medicine, University of Iowa, 431 Newton Road, Iowa City, IA 52442, United States
| | - Prajwal Gurung
- Inflammation Program, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
- Department of Internal Medicine, University of Iowa, 431 Newton Road, Iowa City, IA 52442, United States
- Immunology Graduate Program, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
- Interdisciplinary Graduate Program in Human Toxicology, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
- Center for Immunology and Immune Based Disease, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
- Iowa City Veterans Affairs (VA) Medical Center, 601 US-6, Iowa City, IA 52246, United States
| |
Collapse
|
31
|
Kalafati E, Kastritis E, Bagratuni T. Targeting BCL2 in Waldenström macroglobulinemia: from biology to treatment management. Front Oncol 2025; 15:1564869. [PMID: 40330831 PMCID: PMC12052752 DOI: 10.3389/fonc.2025.1564869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 03/31/2025] [Indexed: 05/08/2025] Open
Abstract
Despite recent advances in the treatment of Waldenström macroglobulenimia (WM), including the development of Bruton tyrosine kinase inhibitors (BTKis), the disease remains incurable highlighting the urgent need for new treatments. The overexpression of BCL2 in WM cells promotes cell survival by resisting apoptosis and contributes to resistance to chemotherapy and targeted therapies. Concurrently, Bcl2 proteins that are encoded by oncogenes supporting cell survival are frequently upregulated in WM, even in the presence of DNA-damaging agents, and hence have emerged as an alternative therapeutic target. Venetoclax serves as a novel orally administered small agent that targets Bcl-2 protein by acting as a BCL2 homology domain 3 (BH3) mimetic and has shown promising results in WM patients, including those previously treated with BTKis. Furthermore, venetoclax, in combination with standard WM regimens, has shown enhanced activity, but further studies are required to elucidate the mechanism of its synergistic action and identify the patients who can benefit from the combined therapy. New BCL2 inhibitors are in advanced stages of clinical development and may offer additional options. The present review will focus on the current knowledge we have on BCL2 inhibitors in WM, the input of these compounds "from bench to bedside," and their utility in managing relapsed/refractory WM patients.
Collapse
Affiliation(s)
| | | | - Tina Bagratuni
- Department of Clinical Therapeutics, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece
| |
Collapse
|
32
|
Chen YT, Lohia GK, Chen S, Liu Z, Wong Fok Lung T, Wang C, Riquelme SA. A host-pathogen metabolic synchrony that facilitates disease tolerance. Nat Commun 2025; 16:3729. [PMID: 40253414 PMCID: PMC12009439 DOI: 10.1038/s41467-025-59134-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 04/10/2025] [Indexed: 04/21/2025] Open
Abstract
Disease tolerance mitigates organ damage from non-resolving inflammation during persistent infections, yet its underlying mechanisms remain unclear. Here we show, in a Pseudomonas aeruginosa pneumonia mouse model, that disease tolerance depends on the mitochondrial metabolite itaconate, which mediates cooperative host-pathogen interactions. In P. aeruginosa, itaconate modifies key cysteine residues in TCA cycle enzymes critical for succinate metabolism, inducing bioenergetic stress and promoting the formation biofilms that are less immunostimulatory and allow the bacteria to integrate into the local microbiome. Itaconate incorporates into the central metabolism of the biofilm, driving exopolysaccharide production-particularly alginate-which amplifies airway itaconate signaling. This itaconate-alginate interplay limits host immunopathology by enabling pulmonary glutamine assimilation, activating glutaminolysis, and thereby restrain detrimental inflammation caused by the inflammasome. Clinical sample analysis reveals that P. aeruginosa adapts to this metabolic environment through compensatory mutations in the anti-sigma-factor mucA, which restore the succinate-driven bioenergetics and disrupt the metabolic synchrony essential for sustaining disease tolerance.
Collapse
Affiliation(s)
- Ying-Tsun Chen
- Department of Pediatrics, Columbia University, New York, NY, USA
| | | | - Samantha Chen
- Department of Pediatrics, Columbia University, New York, NY, USA
| | - Zihua Liu
- Synthetic and Functional Biomolecules Center, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | | | - Chu Wang
- Synthetic and Functional Biomolecules Center, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | | |
Collapse
|
33
|
Li Z, Si P, Meng T, Zhao X, Zhu C, Zhang D, Meng S, Li N, Liu R, Ni T, Yan J, Li H, Zhao N, Zhong C, Qin Y, Chen W, Chen ZJ, Jiao X. CCR8 + decidual regulatory T cells maintain maternal-fetal immune tolerance during early pregnancy. Sci Immunol 2025; 10:eado2463. [PMID: 40249828 DOI: 10.1126/sciimmunol.ado2463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 12/13/2024] [Accepted: 02/18/2025] [Indexed: 04/20/2025]
Abstract
Regulatory T (Treg) cells play a vital role in maintaining maternal immune tolerance to the semiallogeneic fetus during pregnancy. Treg cell population heterogeneity and tissue-specific functions in the human decidua remain largely unknown. Here, using single-cell transcriptomic and T cell receptor sequencing of human CD4+ T cells from first-trimester deciduae and matched peripheral blood of pregnant women, we identified a highly activated, immunosuppressive CCR8+ Treg cell subset specifically enriched in the decidua (dTreg cells). CCR8+ dTreg cells were decreased in patients with recurrent pregnancy loss (RPL) and an abortion-prone mouse model. Depletion of CCR8+ dTreg cells increased susceptibility to fetal loss, with altered decidual immune profiles. Adoptive transfer of CCR8+ Treg cells rescued fetal loss in abortion-prone mice. The CCR8 ligand CCL1 was mainly produced by decidual CD49a+ natural killer cells and was significantly decreased in patients with RPL. Our data demonstrate that CCR8+ dTreg cells are required to maintain maternal-fetal tolerance and highlight potential avenues for RPL therapies.
Collapse
Affiliation(s)
- Zhuqing Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Second Hospital, Shandong University, Jinan, Shandong 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong 250012, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong 250012, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Jinan, Shandong 250012, China
- Department of Reproductive Medicine, Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Pinxin Si
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Second Hospital, Shandong University, Jinan, Shandong 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong 250012, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong 250012, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Jinan, Shandong 250012, China
| | - Tingting Meng
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Second Hospital, Shandong University, Jinan, Shandong 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong 250012, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong 250012, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Jinan, Shandong 250012, China
| | - Xiaoran Zhao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Second Hospital, Shandong University, Jinan, Shandong 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong 250012, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong 250012, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Jinan, Shandong 250012, China
| | - Chendi Zhu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Second Hospital, Shandong University, Jinan, Shandong 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong 250012, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong 250012, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Jinan, Shandong 250012, China
| | - Dunfang Zhang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Shutong Meng
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Second Hospital, Shandong University, Jinan, Shandong 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong 250012, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong 250012, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Jinan, Shandong 250012, China
| | - Nianyu Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Second Hospital, Shandong University, Jinan, Shandong 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong 250012, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong 250012, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Jinan, Shandong 250012, China
| | - Ran Liu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Second Hospital, Shandong University, Jinan, Shandong 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong 250012, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong 250012, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Jinan, Shandong 250012, China
| | - Tianxiang Ni
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Second Hospital, Shandong University, Jinan, Shandong 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong 250012, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong 250012, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Jinan, Shandong 250012, China
| | - Junhao Yan
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Second Hospital, Shandong University, Jinan, Shandong 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong 250012, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong 250012, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Jinan, Shandong 250012, China
| | - Hongchang Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Second Hospital, Shandong University, Jinan, Shandong 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong 250012, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong 250012, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Jinan, Shandong 250012, China
| | - Ning Zhao
- Analytical Biosciences Limited, Beijing 100191, China
| | - Chao Zhong
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Medicine Innovation Center for Fundamental Research on Major Immunology-Related Diseases, Peking University, Beijing 100191, China
| | - Yingying Qin
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Second Hospital, Shandong University, Jinan, Shandong 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong 250012, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong 250012, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Jinan, Shandong 250012, China
- Department of Reproductive Medicine, Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - WanJun Chen
- Mucosal Immunology Section, National Institutes of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892, USA
| | - Zi-Jiang Chen
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Second Hospital, Shandong University, Jinan, Shandong 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong 250012, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong 250012, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Jinan, Shandong 250012, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
- Department of Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xue Jiao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Second Hospital, Shandong University, Jinan, Shandong 250012, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, Shandong 250012, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, Shandong 250012, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, Shandong 250012, China
- Shandong Key Laboratory of Reproductive Research and Birth Defect Prevention, Jinan, Shandong 250012, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No. 2021RU001), Jinan, Shandong 250012, China
| |
Collapse
|
34
|
Lim YJ, Duckworth AD, Clarke K, Kennedy P, Karpha I, Oates M, Gornall M, Kalakonda N, Slupsky JR, Pettitt AR. Influence of polyfunctional Tbet + T cells on specific clinical events in chronic lymphocytic leukaemia. Front Immunol 2025; 16:1528405. [PMID: 40313965 PMCID: PMC12043603 DOI: 10.3389/fimmu.2025.1528405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 03/18/2025] [Indexed: 05/03/2025] Open
Abstract
Introduction T-cell dysfunction is a hallmark of chronic lymphocytic leukemia (CLL), but the extent to which individual CD4+ or CD8+ T-cell subpopulations influence specific clinical events remains unclear. To address this knowledge gap, we utilised high-dimensional mass cytometry to profile circulating CD4+ and CD8+ T-cells in pre-treatment samples from a well-defined cohort of CLL patients undergoing initial therapy as part of a clinical trial. Methods Pre-treatment blood samples from 138 CLL patients receiving initial chemoimmunotherapy containing bendamustine or chlorambucil in the NCRI RIAltO trial (NCT01678430; EudraCT 2011-000919-22) were subjected to deep immunophenotyping by mass cytometry using a bespoke panel of 37 antibodies. T-cell clusters were identified through unsupervised clustering and related to treatment outcomes. Additionally, a randomly selected cohort of 30 CLL patients underwent T-cell stimulation with anti-CD3/CD28 microbeads, followed by cytokine analysis using a separate 36-antibody panel, which included seven cytokines. Results Seventeen CD4+ and 22 CD8+ T-cell clusters were identified in a discovery cohort of 79 patients. Three of these clusters, measured as a proportion of their parental CD4+ or CD8+ populations, correlated with a reduced risk of grade ≥3 infection, grade ≥3 second primary malignancy (SPM) and death, respectively. Three corresponding T-cell subpopulations prospectively defined by non-redundant markers and Boolean gating (ICOS+HLA-DR+PD1+TIGIT+Tbet+CD4+ T-helper cells; CD27+CD28-PD1+Tbet+Eomes+CD8+ cells; and CD27+CD28-GrymB+Tbet+Eomes+CD8+ terminal effector cells) showed the same clinical correlations as the clusters on which they were based. With the exception of SPM for which there were insufficient events, these correlations were confirmed in a separate validation cohort of 59 patients. In-vitro stimulation of a subset of CLL patients in the discovery cohort showed an enrichment of primed and polyfunctional cells in all three Tbet+ T-cell subpopulations of interest. Conclusion Our study provides new insights into the potential for Tbet+ T-cell subpopulations to influence and predict specific clinical events in CLL. This, in turn, raises the possibility that these respective subpopulations could play an important role in controlling infection, solid tumours and CLL itself. Clinical Trial Registration https://www.clinicaltrials.gov/, identifier NCT01678430; https://www.isrctn.com/ISRCTN09988575, identifier EudraCT 2011-000919-22.
Collapse
Affiliation(s)
- Yeong Jer Lim
- Department of Molecular & Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
- Haemato-oncology Department, The Clatterbridge Cancer Centre National Health Service (NHS) Foundation Trust, Liverpool, United Kingdom
| | - Andrew D. Duckworth
- Department of Molecular & Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Kim Clarke
- Computational Biology Facility, University of Liverpool, Liverpool, United Kingdom
| | - Paul Kennedy
- Department of Pharmacology & Therapeutics, University of Liverpool, Liverpool, United Kingdom
| | - Indrani Karpha
- Department of Molecular & Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
- Haemato-oncology Department, The Clatterbridge Cancer Centre National Health Service (NHS) Foundation Trust, Liverpool, United Kingdom
| | - Melanie Oates
- Department of Molecular & Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Matthew Gornall
- Liverpool Clinical Trials Centre, University of Liverpool, Liverpool, United Kingdom
| | - Nagesh Kalakonda
- Department of Molecular & Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
- Haemato-oncology Department, The Clatterbridge Cancer Centre National Health Service (NHS) Foundation Trust, Liverpool, United Kingdom
| | - Joseph R. Slupsky
- Department of Molecular & Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Andrew R. Pettitt
- Department of Molecular & Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
- Haemato-oncology Department, The Clatterbridge Cancer Centre National Health Service (NHS) Foundation Trust, Liverpool, United Kingdom
| |
Collapse
|
35
|
Wang YQ, Wang S, Yi HM, Qian Y, Wang Y, Xu HM, Xu-Monette ZY, Au K, Tian S, Dong Y, Zhao J, Fu D, Mu RJ, Wang SY, Wang L, Young KH, Xu PP, Zhao WL. Practical microenvironment classification in diffuse large B cell lymphoma using digital pathology. Cell Rep Med 2025; 6:102030. [PMID: 40112808 PMCID: PMC12047489 DOI: 10.1016/j.xcrm.2025.102030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 01/15/2025] [Accepted: 02/24/2025] [Indexed: 03/22/2025]
Abstract
Diffuse large B cell lymphoma (DLBCL) is a heterogeneous B cell neoplasm with variable clinical outcomes influenced by both tumor-derived and lymphoma microenvironment (LME) alterations. A recent transcriptomic study identifies four DLBCL subtypes based on LME characteristics: germinal center (GC)-like, mesenchymal (MS), inflammatory (IN), and depleted (DP). However, integrating this classification into clinical practice remains challenging. Here, we utilize deconvolution methods to assess microenvironment component abundance, establishing an LME classification of DLBCL using immunohistochemistry markers and digital pathology based on CD3, CD8, CD68, PD-L1, and collagen. This staining-based algorithm demonstrates over 80% concordance with transcriptome-based classification. Single-cell sequencing confirms that the immune microenvironments distinguished by this algorithm align with transcriptomic profiles. Significant disparities in overall and progression-free survival are observed among LME subtypes following rituximab, cyclophosphamide, doxorubicin, vincristine, and prednisone (R-CHOP) or R-CHOP with targeted agents (R-CHOP-X) immunochemotherapy. LME subtypes differed from distinct immune escape mechanisms, highlighting specific immunotherapeutic targets and supporting application of this classification in future precision medicine trials.
Collapse
Affiliation(s)
- Yu-Qing Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Hematology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shuo Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong-Mei Yi
- Department of Pathology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Qian
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yue Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hai-Min Xu
- Department of Pathology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zijun Y Xu-Monette
- Hematopathology Division and Department of Pathology, Duke University Medical Center, Durham, NC, USA; Duke Cancer Institute, Durham, NC, USA
| | - Kelly Au
- Hematopathology Division and Department of Pathology, Duke University Medical Center, Durham, NC, USA
| | - Shuang Tian
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Dong
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Zhao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Di Fu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rong-Ji Mu
- Department of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shu-Ye Wang
- Department of Hematology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Li Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Pôle de Recherches Sino-Français en Science du Vivant et Génomique, Laboratory of Molecular Pathology, Shanghai, China
| | - Ken H Young
- Hematopathology Division and Department of Pathology, Duke University Medical Center, Durham, NC, USA; Duke Cancer Institute, Durham, NC, USA.
| | - Peng-Peng Xu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Wei-Li Zhao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China; Pôle de Recherches Sino-Français en Science du Vivant et Génomique, Laboratory of Molecular Pathology, Shanghai, China.
| |
Collapse
|
36
|
Wang Q, Cui H, Zhu Y, Zhu Y, He C. Serum Golgi protein 73 (GP73) is a diagnostic and prognostic marker of hepatocellular carcinoma. Front Med (Lausanne) 2025; 12:1571761. [PMID: 40297156 PMCID: PMC12034698 DOI: 10.3389/fmed.2025.1571761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 03/31/2025] [Indexed: 04/30/2025] Open
Abstract
Introduction Hepatocellular carcinoma (HCC) constitutes a significant global health burden, and is characterized by limited early detection methods and poor survival rates. Golgi protein 73 (GP73), previously associated with liver-related diseases, has a controversial diagnostic value for HCC. The present study aimed to determine the diagnostic efficacy of serum GP73 (sGP73) levels in HCC and to explore their potential correlations with the development of HCC. Methods The levels of sGP73 and serum alpha-fetoprotein (sAFP) were measured in 134 HCC patients, 200 healthy controls (HCs), and 45 non-HCC patients with various liver diseases. Additionally, immunohistochemical staining was conducted on paraffin-embedded tissue samples obtained from 30 HCC patients to examine the expression of CD4+ T cells, CD8+ T cells, Foxp3+ Treg cells, Ki-67, and interferon-gamma (IFN-γ) in the tissue specimens. Results sGP73 and sAFP were markedly higher in HCC patients than in HCs and non-HCC patients. However, sGP73 showed significantly higher sensitivity as a diagnostic marker for HCC than sAFP. The combination of sGP73 and sAFP further improved the accuracy (AUROC: 0.830). Besides, in the immunohistochemical staining analyses, sGP73-positive patients had lower expression of CD4+ and CD8+ T cells, higher expression of Foxp3+ Treg cells, higher expression of nuclear Ki67, and lower expression of IFN-γ than GP73-negative patients. In addition, sGP73-positive patients tended to have higher mortality rate, higher rate of metastasis, higher AFP levels, and more pronounced liver inflammation and damage than GP73-negative patients. Conclusions sGP73 could be utilized as a marker for the diagnosis of HCC, and may be implicated in the development of HCC through its interactions with the tumor microenvironment.
Collapse
Affiliation(s)
- Qian Wang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Hongxia Cui
- Department of Pathology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yaping Zhu
- Department of Clinical Laboratory, Kunshan Hospital of Chinese Medicine, Kunshan, Jiangsu, China
| | - Yichun Zhu
- Department of Clinical Laboratory, Kunshan Hospital of Chinese Medicine, Kunshan, Jiangsu, China
| | - Chunyan He
- Department of Clinical Laboratory, Kunshan Hospital of Chinese Medicine, Kunshan, Jiangsu, China
| |
Collapse
|
37
|
Fu L, Upadhyay R, Pokrovskii M, Chen FM, Romero-Meza G, Griesemer A, Littman DR. PRDM16-dependent antigen-presenting cells induce tolerance to gut antigens. Nature 2025:10.1038/s41586-025-08982-4. [PMID: 40228524 DOI: 10.1038/s41586-025-08982-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 04/04/2025] [Indexed: 04/16/2025]
Abstract
The gastrointestinal tract is continuously exposed to foreign antigens in food and commensal microorganisms with potential to induce adaptive immune responses. Peripherally induced T regulatory (pTreg) cells are essential for mitigating inflammatory responses to these agents1-4. Although RORγt+ antigen-presenting cells (APCs) have been shown to programme gut microbiota-specific pTreg cells5-7, their definition remains incomplete, and the APC responsible for food tolerance has remained unknown. Here we identify an APC subset that is required for differentiation of both food- and microbiota-specific pTreg cells and for establishment of oral tolerance. Development and function of these APCs require expression of the transcription factors PRDM16 and RORγt, as well as a unique Rorc(t) cis-regulatory element. Gene expression, chromatin accessibility, and surface marker analysis establish the pTreg-inducing APCs as myeloid in origin, distinct from type 3 innate lymphoid cells, and sharing epigenetic profiles with classical dendritic cells, and designate them PRDM16+RORγt+ tolerizing dendritic cells (tolDCs). Upon genetic perturbation of tolDCs, we observe a substantial increase in food antigen-specific T helper 2 cells in lieu of pTreg cells, leading to compromised tolerance in mouse models of asthma and food allergy. Single-cell analyses of freshly resected mesenteric lymph nodes from a human organ donor, as well as multiple specimens of human intestine and tonsil, reveal candidate tolDCs with co-expression of PRDM16 and RORC and an extensive transcriptome shared with tolDCs from mice, highlighting an evolutionarily conserved role across species. Our findings suggest that a better understanding of how tolDCs develop and how they regulate T cell responses to food and microbial antigens could offer new insights into developing therapeutic strategies for autoimmune and allergic diseases as well as organ transplant tolerance.
Collapse
Affiliation(s)
- Liuhui Fu
- Department of Cell Biology, New York University School of Medicine, New York, NY, USA
| | - Rabi Upadhyay
- Department of Cell Biology, New York University School of Medicine, New York, NY, USA
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Maria Pokrovskii
- Department of Cell Biology, New York University School of Medicine, New York, NY, USA
- Calico Life Sciences, South San Francisco, CA, USA
| | - Francis M Chen
- Department of Cell Biology, New York University School of Medicine, New York, NY, USA
| | - Gabriela Romero-Meza
- Department of Cell Biology, New York University School of Medicine, New York, NY, USA
- Howard Hughes Medical Institute, New York, NY, USA
| | - Adam Griesemer
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
| | - Dan R Littman
- Department of Cell Biology, New York University School of Medicine, New York, NY, USA.
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA.
- Howard Hughes Medical Institute, New York, NY, USA.
| |
Collapse
|
38
|
Xiao Q, Xu P, Xu W, Song Q, Mao Y. Identification of characteristic genes in cutaneous squamous cell carcinoma based on weighted gene co-expression network analysis. Front Genet 2025; 16:1470584. [PMID: 40296873 PMCID: PMC12034687 DOI: 10.3389/fgene.2025.1470584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 03/31/2025] [Indexed: 04/30/2025] Open
Abstract
Objective This study aims to identify characteristic genes associated with cutaneous squamous cell carcinoma (cSCC). Methods Differentially expressed genes (DEGs) and hub genes in key module were identified using the limma package and weighted gene co-expression network analysis (WGCNA) in R software, respectively. The intersection of these genes was then subjected to LASSO regression to pinpoint characteristic genes. The correlation between immune cell infiltration and these characteristic genes was further elucidated using single-sample Gene Set Enrichment Analysis and Spearman correlation analysis. Results A total of 113 DEGs were identified, along with their associated biological pathways. From this pool, five characteristic genes-ADH1B, CCL27, ID4, LRP4 and S100A9-were selected and validated. Immune infiltration analysis revealed significant correlations between these genes and various immune cell types, particularly with CCL27, ID4, LRP4 and S100A9. Conclusion The identification of characteristic genes for cSCC provides valuable insights into its molecular mechanisms. The correlations between these genes and immune cell infiltration suggests their potential roles in tumor immunity.
Collapse
Affiliation(s)
- Qipeng Xiao
- Department of Dermatology, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi, China
| | - Pengfei Xu
- Department of Dermatology, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi, China
| | - Wenjun Xu
- College of Nursing, Jiujiang Vocational University, Jiujiang, Jiangxi, China
| | - Qiuhe Song
- Department of Dermatology, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi, China
| | - Yousheng Mao
- Department of Dermatology, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi, China
- Jiujiang Clinical Precision Medicine Research Center, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi, China
| |
Collapse
|
39
|
Pan Y, Zhou H, Sun Z, Zhu Y, Zhang Z, Han J, Liu Y, Wang Q. Regulatory T cells in solid tumor immunotherapy: effect, mechanism and clinical application. Cell Death Dis 2025; 16:277. [PMID: 40216744 PMCID: PMC11992189 DOI: 10.1038/s41419-025-07544-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 01/12/2025] [Accepted: 03/17/2025] [Indexed: 04/14/2025]
Abstract
The tumor-immune response is mobilized to suppress tumorigenesis, while the immune microenvironment and lymph node microenvironment are formed gradually during tumor progression. In fact, tumor surface antigens are not easily recognized by antigen-presenting cells. So it is hard for the immune system to kill the newly formed tumor cells effectively. In a normal immune environment, immune function is always suppressed to maintain the stability of the body, and regulatory T cells play an important role in maintaining immune suppression. However, during tumorigenesis, the suppression of regulatory T cell immune functions is more likely to contribute to tumor cell proliferation and migration leading directly to tumor progression. Therefore, focusing on the role of regulatory T cells in tumor immunity could improve tumor immunotherapy outcomes in the clinic. Regulatory T cells are more mature in hematologic system tumors than in solid tumors. However, there are continuing efforts to apply regulatory T cells for immunotherapy in solid tumors. This review describes the role of regulatory T cells in solid tumor immunotherapy from the perspective of prognosis, immune microenvironment remodeling, and current clinical applications. This summary could help us better understand the mechanisms of regulatory T cells in solid tumor immunotherapy and further expand their clinical application.
Collapse
Affiliation(s)
- Yan Pan
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
- Institute of Cancer Research, Henan Academy of Innovations in Medical Science, Zhengzhou, 451162, China
| | - Hanqiong Zhou
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
- Institute of Cancer Research, Henan Academy of Innovations in Medical Science, Zhengzhou, 451162, China
| | - Zhenqiang Sun
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450001, China
| | - Yichen Zhu
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
- Institute of Cancer Research, Henan Academy of Innovations in Medical Science, Zhengzhou, 451162, China
| | - Zhe Zhang
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
- Institute of Cancer Research, Henan Academy of Innovations in Medical Science, Zhengzhou, 451162, China
| | - Jing Han
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
- Institute of Cancer Research, Henan Academy of Innovations in Medical Science, Zhengzhou, 451162, China
| | - Yang Liu
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China.
| | - Qiming Wang
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China.
- Institute of Cancer Research, Henan Academy of Innovations in Medical Science, Zhengzhou, 451162, China.
| |
Collapse
|
40
|
Masmoudi D, Villalba M, Alix-Panabières C. Natural killer cells: the immune frontline against circulating tumor cells. J Exp Clin Cancer Res 2025; 44:118. [PMID: 40211394 PMCID: PMC11983744 DOI: 10.1186/s13046-025-03375-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 03/25/2025] [Indexed: 04/14/2025] Open
Abstract
Natural killer (NK) play a key role in controlling tumor dissemination by mediating cytotoxicity towards cancer cells without the need of education. These cells are pivotal in eliminating circulating tumor cells (CTCs) from the bloodstream, thus limiting cancer spread and metastasis. However, aggressive CTCs can evade NK cell surveillance, facilitating tumor growth at distant sites. In this review, we first discuss the biology of NK cells, focusing on their functions within the tumor microenvironment (TME), the lymphatic system, and circulation. We then examine the immune evasion mechanisms employed by cancer cells to inhibit NK cell activity, including the upregulation of inhibitory receptors. Finally, we explore the clinical implications of monitoring circulating biomarkers, such as NK cells and CTCs, for therapeutic decision-making and emphasize the need to enhance NK cell-based therapies by overcoming immune escape mechanisms.
Collapse
Affiliation(s)
- Doryan Masmoudi
- Laboratory of Rare Circulating Human Cells, University Medical Center of Montpellier, Montpellier, France
| | - Martin Villalba
- IRMB, Univ Montpellier, INSERM, CHU Montpellier, CNRS, Montpellier, France
| | - Catherine Alix-Panabières
- Laboratory of Rare Circulating Human Cells, University Medical Center of Montpellier, Montpellier, France.
- CREEC/CANECEV, MIVEGEC (CREES), University of Montpellier, CNRS, Montpellier, IRD, France.
- European Liquid Biopsy Society (ELBS), Hamburg, Germany.
- LCCRH, Site Unique de Biologie (SUB), 641, Avenue du Doyen Gaston Giraud, Montpellier, 34093, France.
| |
Collapse
|
41
|
Moës B, Krueger J, Kazanova A, Liu C, Gao Y, Ponnoor NA, Castoun-Puckett L, Lazo ACO, Huong L, Cabald AL, Tu TH, Rudd CE. GSK-3 regulates CD4-CD8 cooperation needed to generate super-armed CD8+ cytolytic T cells against tumors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.08.642085. [PMID: 40161618 PMCID: PMC11952298 DOI: 10.1101/2025.03.08.642085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
While immune checkpoint blockade (ICB) has revolutionized cancer treatment, the key T-cell signaling pathways responsible for its potency remain unclear. GSK-3 is an inhibitory kinase that is most active in resting T-cells. In this study, we demonstrate that GSK-3 facilitates PD-1 blockade, an effect seen by modulating CD4 T-cell help for CD8+ CTL responses against ICB resistant tumors. We show that GSK-3 controls metabolic reprogramming towards glycolysis and synergizes with PD-1 to induce a transcriptional program that reduces suppressive CD4+ Treg numbers while generating super-armed effector-memory CD8+ CTLs that express an unprecedented 7/9 granzymes from the genome. Crucially, we found that GSK-3 cooperates with PD-1 blockade to determine the dependency of CD8+ CTLs on help from CD4+ T-cells. Our study unravels a novel cooperative PD-1 blockade-dependent signaling pathway that potentiates CTL responses against tumors, offering a new strategy to overcome immunotherapy resistance by modulating CD4+ helper and CD8+ cytotoxic functions. Significance This study demonstrates for the first time that GSK-3 controls the crosstalk between CD4+ and CD8+ T cells, synergizing with anti-PD-1 therapy to overcome resistance to checkpoint blockade and to generate super-armed CD8+ effector cells in cancer immunotherapy. This newly uncovered GSK-3-dependent CD4-CD8 T-cell crosstalk mechanism presents a new approach to enhance anti-PD-1 immunotherapy.
Collapse
|
42
|
Tang L, Zhang J, Oumata N, Mignet N, Sollogoub M, Zhang Y. Sialyl Lewis X (sLe x):Biological functions, synthetic methods and therapeutic implications. Eur J Med Chem 2025; 287:117315. [PMID: 39919437 DOI: 10.1016/j.ejmech.2025.117315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 01/20/2025] [Accepted: 01/20/2025] [Indexed: 02/09/2025]
Abstract
Carbohydrates are shown to be crucial to several biological processes. They are essential mediators of cell-cell recognition processes. Among them, Sialyl Lewis X (sLex) is a very significant structure in the human body. It is a critical tetrasaccharide that plays a pivotal role in various biological processes, including cell adhesion, immune response, and cancer metastasis. Known as the blood group antigen, sLex is also referred to as cluster of differentiation 15s (CD15s) or stage-specific embryonic antigen 1 (SSEA-1). sLex is not only a prominent blood group antigen, but also involved in the attraction of sperm to the egg during fertilization, prominently displayed at the terminus of glycolipids on the cell surface. By describing the synthetic methods and biological functions of sLex, this review underscores the importance of sLex in both fundamental and applied sciences and its potential to impact clinical practice.
Collapse
Affiliation(s)
- Leyu Tang
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire, UMR 8232, 4 Place Jussieu, 75005, Paris, France
| | - Jiaxu Zhang
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire, UMR 8232, 4 Place Jussieu, 75005, Paris, France
| | - Nassima Oumata
- Université Paris Cité, UCTBS, Inserm U 1267, CNRS, UMR 8258, 4 Avenue de l'Observatoire, 75006, Paris, France
| | - Nathalie Mignet
- Université Paris Cité, UCTBS, Inserm U 1267, CNRS, UMR 8258, 4 Avenue de l'Observatoire, 75006, Paris, France
| | - Matthieu Sollogoub
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire, UMR 8232, 4 Place Jussieu, 75005, Paris, France
| | - Yongmin Zhang
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire, UMR 8232, 4 Place Jussieu, 75005, Paris, France; Fuyang Institute & School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 311422, Zhejiang, China; Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, 571158, China.
| |
Collapse
|
43
|
Meng X, Zhu Y, Liu K, Wang Y, Liu X, Liu C, Zeng Y, Wang S, Gao X, Shen X, Chen J, Tao S, Xu Q, Dong L, Shen L, Wang L. CXXC-finger protein 1 associates with FOXP3 to stabilize homeostasis and suppressive functions of regulatory T cells. eLife 2025; 13:RP103417. [PMID: 40183773 PMCID: PMC11970909 DOI: 10.7554/elife.103417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025] Open
Abstract
FOXP3-expressing regulatory T (Treg) cells play a pivotal role in maintaining immune homeostasis and tolerance, with their activation being crucial for preventing various inflammatory responses. However, the mechanisms governing the epigenetic program in Treg cells during their dynamic activation remain unclear. In this study, we demonstrate that CXXC-finger protein 1 (CXXC1) interacts with the transcription factor FOXP3 and facilitates the regulation of target genes by modulating H3K4me3 deposition. Cxxc1 deletion in Treg cells leads to severe inflammatory disease and spontaneous T cell activation, with impaired immunosuppressive function. As a transcriptional regulator, CXXC1 promotes the expression of key Treg functional markers under steady-state conditions, which are essential for the maintenance of Treg cell homeostasis and their suppressive functions. Epigenetically, CXXC1 binds to the genomic regulatory regions of Treg program genes in mouse Treg cells, overlapping with FOXP3-binding sites. Given its critical role in Treg cell homeostasis, CXXC1 presents itself as a promising therapeutic target for autoimmune diseases.
Collapse
Affiliation(s)
- Xiaoyu Meng
- Institute of Immunology and Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical CenterHangzhouChina
| | - Yezhang Zhu
- Department of Hematology, Tongji Hospital, School of Medicine, Tongji UniversityShanghaiChina
- Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center of Stem Cell Research, National Stem Cell Translational Resource Center, School of Life Sciences and Technology, Tongji UniversityShanghaiChina
| | - Kuai Liu
- Institute of Immunology and Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical CenterHangzhouChina
| | - Yuxi Wang
- Laboratory Animal Center, Zhejiang UniversityHangzhouChina
| | - Xiaoqian Liu
- Institute of Immunology and Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical CenterHangzhouChina
| | - Chenxin Liu
- Zhejiang University School of MedicineHangzhouChina
| | - Yan Zeng
- Institute of Immunology and Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical CenterHangzhouChina
| | - Shuai Wang
- Institute of Immunology and Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical CenterHangzhouChina
| | - Xianzhi Gao
- Institute of Immunology and Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical CenterHangzhouChina
| | - Xin Shen
- Co-Facility Center, Zhejiang University School of MedicineHangzhouChina
| | - Jing Chen
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Sijue Tao
- Laboratory Animal Center, Zhejiang UniversityHangzhouChina
| | - Qianying Xu
- Zhejiang University School of MedicineHangzhouChina
| | - Linjia Dong
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical CollegeHangzhouChina
| | - Li Shen
- MOE Key Laboratory of Biosystems Homeostasis & Protection and Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang UniversityHangzhouChina
- Department of Orthopedics Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhouChina
| | - Lie Wang
- Institute of Immunology and Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical CenterHangzhouChina
- Laboratory Animal Center, Zhejiang UniversityHangzhouChina
- Future Health Laboratory, Innovation Center of Yangtze River Delta, Zhejiang UniversityJiaxingChina
| |
Collapse
|
44
|
Ryba-Stanisławowska M. Unraveling Th subsets: insights into their role in immune checkpoint inhibitor therapy. Cell Oncol (Dordr) 2025; 48:295-312. [PMID: 39325360 PMCID: PMC11996958 DOI: 10.1007/s13402-024-00992-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2024] [Indexed: 09/27/2024] Open
Abstract
T helper (Th) cell subsets play pivotal roles in regulating immune responses within the tumor microenvironment, influencing both tumor progression and anti-tumor immunity. Among these subsets, Th1 cells promote cytotoxic responses through the production of IFN-γ, while Th2 cells and regulatory T cells (Tregs) exert immunosuppressive effects that support tumor growth. Th9 and Th17 cells have context-dependent roles, contributing to both pro-inflammatory and regulatory processes in tumor immunity. Tumor antigen-specific T cells within the tumor microenvironment often exhibit a dysfunctional phenotype due to increased expression of inhibitory receptors such as CTLA-4 and PD-1, leading to reduced antitumor activity. Monoclonal antibodies that block these inhibitory signals-collectively known as immune checkpoint inhibitors (ICIs)-can reactivate these T cells, enhancing their ability to target and destroy cancer cells. Recent advancements have highlighted the critical role of T helper subsets in modulating responses to ICIs, with their interactions remaining a focus of ongoing research. Both positive and negative effects of ICIs have been reported in relation to Th cell subsets, with some effects depending on the type of tumor microenvironment. This review summarizes the crucial roles of different T helper cell subsets in tumor immunity and their complex relationship with immune checkpoint inhibitor therapy.
Collapse
Affiliation(s)
- Monika Ryba-Stanisławowska
- Department of Medical Immunology, Faculty of Medicine, Medical University of Gdańsk, Dębinki 1, Gdańsk, 80-211, Poland.
| |
Collapse
|
45
|
Stüve P, Godoy GJ, Ferreyra FN, Hellriegel F, Boukhallouk F, Kao YS, More TH, Matthies AM, Akimova T, Abraham WR, Kaever V, Schmitz I, Hiller K, Lochner M, Salomon BL, Beier UH, Rehli M, Sparwasser T, Berod L. ACC1 is a dual metabolic-epigenetic regulator of Treg stability and immune tolerance. Mol Metab 2025; 94:102111. [PMID: 39929287 PMCID: PMC11893314 DOI: 10.1016/j.molmet.2025.102111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 02/06/2025] [Indexed: 02/17/2025] Open
Abstract
OBJECTIVE Regulatory T cells (Tregs) are essential in maintaining immune tolerance and controlling inflammation. Treg stability relies on transcriptional and post-translational mechanisms, including histone acetylation at the Foxp3 locus and FoxP3 protein acetylation. Additionally, Tregs depend on specific metabolic programs for differentiation, yet the underlying molecular mechanisms remain elusive. We aimed to investigate the role of acetyl-CoA carboxylase 1 (ACC1) in the differentiation, stability, and function of regulatory T cells (Tregs). METHODS We used either T cell-specific ACC1 knockout mice or ACC1 inhibition via a pharmacological agent to examine the effects on Treg differentiation and stability. The impact of ACC1 inhibition on Treg function was assessed in vivo through adoptive transfer models of Th1/Th17-driven inflammatory diseases. RESULTS Inhibition or genetic deletion of ACC1 led to an increase in acetyl-CoA availability, promoting enhanced histone and protein acetylation, and sustained FoxP3 transcription even under inflammatory conditions. Mice with T cell-specific ACC1 deletion exhibited an enrichment of double positive RORγt+FoxP3+ cells. Moreover, Tregs treated with an ACC1 inhibitor demonstrated superior long-term stability and an enhanced capacity to suppress Th1/Th17-driven inflammatory diseases in adoptive transfer models. CONCLUSIONS We identified ACC1 as a metabolic checkpoint in Treg biology. Our data demonstrate that ACC1 inhibition promotes Treg differentiation and long-term stability in vitro and in vivo. Thus, ACC1 serves as a dual metabolic and epigenetic hub, regulating immune tolerance and inflammation by balancing de novo lipid synthesis and protein acetylation.
Collapse
Affiliation(s)
- Philipp Stüve
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, Germany; A Joint Venture Between the Hannover Medical School (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover 30625, Germany; Leibniz Institute for Immunotherapy, Regensburg, Germany; Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz 55122, Germany
| | - Gloria J Godoy
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz 55131, Germany
| | - Fernando N Ferreyra
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz 55131, Germany; Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina; Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Florencia Hellriegel
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz 55131, Germany; Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina; Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Fatima Boukhallouk
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz 55122, Germany
| | - Yu-San Kao
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz 55122, Germany
| | - Tushar H More
- Department of Bioinformatics and Biochemistry, BRICS, Technische Universität Braunschweig, 38106 Braunschweig, Germany
| | - Anne-Marie Matthies
- Systems-Oriented Immunology and Inflammation Research Group, Department of Experimental Immunology, HZI, Braunschweig 38124, Germany; Institute for Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, Magdeburg 39106, Germany; Institute for Molecular Immunology, Ruhr-University Bochum, Bochum 44801, Germany
| | - Tatiana Akimova
- Division of Transplant Immunology, Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wolf-Rainer Abraham
- Department of Bioinformatics and Biochemistry, BRICS, Technische Universität Braunschweig, 38106 Braunschweig, Germany; Department of Chemical Microbiology, HZI, Braunschweig 38124, Germany
| | - Volkhard Kaever
- Research Core Unit Metabolomics, MHH, Hannover 30625, Germany
| | - Ingo Schmitz
- Systems-Oriented Immunology and Inflammation Research Group, Department of Experimental Immunology, HZI, Braunschweig 38124, Germany; Institute for Molecular and Clinical Immunology, Otto-von-Guericke University Magdeburg, Magdeburg 39106, Germany; Institute for Molecular Immunology, Ruhr-University Bochum, Bochum 44801, Germany
| | - Karsten Hiller
- Department of Bioinformatics and Biochemistry, BRICS, Technische Universität Braunschweig, 38106 Braunschweig, Germany
| | - Matthias Lochner
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, Germany; A Joint Venture Between the Hannover Medical School (MHH) and the Helmholtz Centre for Infection Research (HZI), Hannover 30625, Germany; Institute of Medical Microbiology and Hospital Epidemiology, MHH, Hannover 30625, Germany
| | - Benoît L Salomon
- Sorbonne Université, INSERM, CNRS, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris 75013, France
| | - Ulf H Beier
- Division of Nephrology and Department of Pediatrics, Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael Rehli
- Leibniz Institute for Immunotherapy, Regensburg, Germany; Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Tim Sparwasser
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz 55122, Germany; Research Center for Immunotherapy (FZI), University Medical Center Mainz, 55131 Mainz, Germany
| | - Luciana Berod
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz 55131, Germany; Research Center for Immunotherapy (FZI), University Medical Center Mainz, 55131 Mainz, Germany.
| |
Collapse
|
46
|
Wu L, Zhu L, Chen J. Diverse potential of chimeric antigen receptor-engineered cell therapy: Beyond cancer. Clin Transl Med 2025; 15:e70306. [PMID: 40205818 PMCID: PMC11982526 DOI: 10.1002/ctm2.70306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 03/24/2025] [Accepted: 03/31/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND Chimeric antigen receptor (CAR)-engineered cell therapies have made significant progress in haematological cancer treatment. This success has motivated researchers to investigate its potential applications in non-cancerous diseases, with substantial strides already made in this field. MAIN BODY This review summarises the latest research on CAR-engineered cell therapies, with a particular focus on CAR-T cell therapy for non-cancerous diseases, including but not limited to infectious diseases, autoimmune diseases, cardiac diseases and immune-mediated disorders in transplantation. Additionally, the review discusses the current obstacles that need to be addressed for broader clinical applications. CONCLUSION With ongoing research and continuous improvements, CAR-engineered cell therapy holds promise as a potent tool for treating various diseases in the future. KEY POINTS CAR-engineered cell therapy has expanded beyond cancer to treat autoimmune diseases, infections, cardiac diseases, and transplant-related rejection. The CAR platform is diverse, with various cell types such as CAR-T, CAR-NK, and CAR-M potentially suited for different disease contexts. The safety, efficacy, and practicality of CAR cell therapy in non-cancer diseases remain challenging, requiring further technological optimization and clinical translation.
Collapse
Affiliation(s)
- Lvying Wu
- Institute of Clinical MedicineThe Second Affiliated Hospital of Hainan Medical UniversityHaikouHainanChina
| | - Lingfeng Zhu
- Minimally Invasive Urology and Translational Medicine CenterFuzhou First General Hospital Affiliated With Fujian Medical UniversityFuzhouFujianChina
| | - Jin Chen
- Institute of Clinical MedicineThe Second Affiliated Hospital of Hainan Medical UniversityHaikouHainanChina
- Minimally Invasive Urology and Translational Medicine CenterFuzhou First General Hospital Affiliated With Fujian Medical UniversityFuzhouFujianChina
| |
Collapse
|
47
|
Valentín-Quiroga J, Zarauza-Santoveña A, López-Collazo E, Ferreira LMR. Chimeric anti-HLA antibody receptor engineered human regulatory T cells suppress alloantigen-specific B cells from pre-sensitized transplant recipients. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.27.645777. [PMID: 40236118 PMCID: PMC11996358 DOI: 10.1101/2025.03.27.645777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Organ transplantation is a lifesaving procedure, with 50,000 transplants happening every year in the United States. However, many patients harbor antibodies and B cells directed against allogeneic human leukocyte antigen (HLA) molecules, notably HLA-A2, greatly decreasing their likelihood of receiving a compatible organ. Moreover, antibody-mediated rejection is a significant contributor to chronic transplant rejection. Current strategies to desensitize patients non- specifically target circulating antibodies and B cells, resulting in poor efficacy and complications. Regulatory T cells (Tregs) are immune cells dedicated to suppressing specific immune responses by interacting with both innate and adaptive immune cells. Here, we genetically modified human Tregs with a chimeric anti-HLA antibody receptor (CHAR) consisting of an extracellular HLA-A2 protein fused to a CD28-CD3zeta intracellular signaling domain, driving Treg activation upon recognition of anti-HLA-A2 antibodies on the surface of alloreactive B cells. We find that HLA-A2 CHAR Tregs get activated specifically by anti-HLA-A2 antibody-producing cells. Of note, HLA-A2 CHAR activation does not negatively affect Treg stability, as measured by expression of the Treg lineage transcription factors FOXP3 and HELIOS. Interestingly, HLA-A2 CHAR Tregs are not cytotoxic towards anti-HLA-A2 antibody-producing cells, unlike HLA-A2 CHAR modified conventional CD4 + T cells. Importantly, HLA-A2 CHAR Tregs recognize and significantly suppress high affinity IgG antibody production by B cells from HLA-A2 sensitized patients. Altogether, our results provide proof-of-concept of a new strategy to specifically inhibit alloreactive B cells to desensitize transplant recipients.
Collapse
|
48
|
Lee S, Dohlman TH, Dana R. Immunology in corneal transplantation-From homeostasis to graft rejection. Transplant Rev (Orlando) 2025; 39:100909. [PMID: 39798206 PMCID: PMC11975484 DOI: 10.1016/j.trre.2025.100909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 12/19/2024] [Accepted: 01/08/2025] [Indexed: 01/15/2025]
Abstract
Immunology depends on maintaining a delicate balance within the human body, and disruptions can result in conditions such as autoimmune diseases, immunodeficiencies, and hypersensitivity reactions. This balance is especially crucial in transplantation immunology, where one of the primary challenges is preventing graft rejection. Such rejection can lead to organ failure, increased patient mortality, and higher healthcare costs due to the limited availability of donor tissues relative to patient needs. Xenotransplantation, like using porcine corneas for human transplants, offers a potential solution to the donor tissue shortage but faces substantial immunological rejection issues. To prevent rejection in both allo- and xenotransplantation, a deep understanding of how the body maintains immunological balance is essential, particularly since achieving tolerance to non-self tissues is considered the "holy grail" of the field. The cornea, the most frequently transplanted solid organ, has a high acceptance rate due to its immune-privileged status and serves as an ideal model for studying graft rejection mechanisms that disrupt tolerance. However, multiple immune pathways complicate our understanding of these mechanisms. This review examines the rejection mechanisms in corneal transplantation, identifying key cells involved and potential therapeutic strategies to induce and maintain immunological tolerance in both allo- and xenografts across various transplants.
Collapse
Affiliation(s)
- Seokjoo Lee
- Laboratory of Ocular Immunology, Transplantation, and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Thomas H Dohlman
- Laboratory of Ocular Immunology, Transplantation, and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Reza Dana
- Laboratory of Ocular Immunology, Transplantation, and Regeneration, Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
49
|
Jin F, Gridley J, Kumari A, Saeidi A, Holland B, Elrod E, Dravid P, Trivedi S, Kapoor A, Thapa M, Grakoui A. Hyperfunctional T cell responses unchecked by regulatory T cells are unable to resolve hepaciviral infection without humoral contribution. J Hepatol 2025; 82:604-614. [PMID: 39423870 PMCID: PMC11911089 DOI: 10.1016/j.jhep.2024.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 09/19/2024] [Accepted: 10/03/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND & AIMS The most recent T cell-based vaccine against HCV tested in humans failed to swing the pendulum from chronicity to resolution, despite eliciting cellular responses in most individuals. These results naturally evoke the question of whether hyperactivated responses of a single adaptive immune arm are capable of inducing HCV clearance or if coordinated efforts between antibodies and T cells are indeed necessary. Here, we sought to address this point in determining whether the suppression of antiviral T cell and IgG responses by regulatory T cells (Tregs) is a critical prerequisite of delayed viral clearance or overt chronicity. METHODS Using a surrogate model of HCV infection, rodent hepacivirus (RHV) infection in mice, we utilized Foxp3-DTR mice to assess how Tregs modulate the generation of acute antiviral adaptive immune responses and indirectly dictate infection fate via intracellular flow cytometry staining, ELISA, RNA sequencing, and qPCR. RESULTS Transient depletion of Tregs prior to infection decreased viral-specific CD4+ T cell function, IgG production, and delayed viral clearance. In contrast, transient Treg depletion after infection increased both T cell functionality and IgG production, thereby facilitating accelerated viral clearance. Hyperactivated T cells, achieved via transient Treg depletion, were unable to clear the virus as an isolated effector arm without the help of viral-specific IgG production. CONCLUSIONS Tregs control the outcome of RHV infection via direct modulation of CD4+ T cells and IgG production. Hyperactivated T cell responses are incapable of compensating for experimentally induced lack of antibodies, further reinforcing the notion of cooperative interplay between adaptive immune arms in facilitating hepaciviral clearance. IMPACT AND IMPLICATIONS Herein, we demonstrate how timing of regulatory T cell depletion determines the fate of effector T cells, humoral responses, and the kinetics of viral clearance. Our observations provide direct evidence that functional T cell responses are incapable of compensating for suboptimal humoral responses to facilitate viral resolution. Our results imply that future HCV vaccine regimens should not solely rely on eliciting focused responses of a single effector arm, but rather incorporate immunogens capable of inducing durable features of both humoral and cellular memory.
Collapse
Affiliation(s)
- Fengzhi Jin
- Emory National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - John Gridley
- Emory National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Anuradha Kumari
- Emory National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Alireza Saeidi
- Emory National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Brantley Holland
- Emory National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Elizabeth Elrod
- Emory National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Piyush Dravid
- Center for Vaccines and Immunity, The Research Institute at Nationwide Children's Hospital and Department of Pediatrics, Ohio State University, Columbus, Ohio, USA
| | - Sheetal Trivedi
- Center for Vaccines and Immunity, The Research Institute at Nationwide Children's Hospital and Department of Pediatrics, Ohio State University, Columbus, Ohio, USA
| | - Amit Kapoor
- Center for Vaccines and Immunity, The Research Institute at Nationwide Children's Hospital and Department of Pediatrics, Ohio State University, Columbus, Ohio, USA
| | - Manoj Thapa
- Emory National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Arash Grakoui
- Emory National Primate Research Center, Emory University, Atlanta, Georgia, USA; Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA.
| |
Collapse
|
50
|
Balçık OY, Yılmaz F. FOXP3/TLS; a prognostic marker in patients with bladder carcinoma without muscle invasion. Urol Oncol 2025; 43:268.e9-268.e26. [PMID: 39668105 DOI: 10.1016/j.urolonc.2024.11.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 11/04/2024] [Accepted: 11/12/2024] [Indexed: 12/14/2024]
Abstract
OBJECTIVE Bladder carcinoma (BC) is a common type of cancer. Approximately 20% of BC patients have non-muscle invasive bladder cancer (NMIBC). Despite adequate BCG treatment, recurrence occurs in approximately 40% of the patients. There is no adequate prognostic marker for recurrence in a group of patients. Forkhead box P3 (FOXP3) is a regulatory T cell marker that sometimes exhibits anti-tumoral effects and can be used as a tumor marker. T-cell immunoglobulin and mucin domain 3 (TIM-3) is an immune checkpoint inhibitor of T cells. Tertiary lymphoid structures (TLS) increase malignancy and inflammation in non-lymphoid organs. Therefore, we aimed to evaluate the prognostic value of FOXP3, TIM-3, and TLS in patients with NMIBC. METHODS Patients with pathologically confirmed NMIBC were included in this study. Stromal and intraepithelial cells were evaluated separately using immunohistochemistry, and FOXP3, TIM-3, TLS, FOXP3/TLS, and TIM-3/TLS were calculated and noted. The cutoff value was determined using ROC analysis. Recurrence-free survival (RFS) and overall survival (OS) were evaluated using univariate and multivariate Cox proportional hazard analyses. RESULTS The study included ninety-six patients. FOXP3/TLS high group had a better RFS than FOXP3/TLS low group (P = 0.001; HR, 0.079; 95% CI, 0.019-0.337). This was also significant in the multivariate analysis (P = 0.018; HR, 0.125; 95% CI, 0.022-0.705). In the group receiving BCG, FOXP3/TLS, FOXP3-TLS, TIM-3-TLS and TIM-3/TLS elevation were lower in patients with relapse than in patients without relapse and were statistically significant. Combined TIM-3 and FOXP3 elevation was found to be good prognostic regardless of whether it was found in intraepithelial, stromal or TLS. CONCLUSION FOXP3/TLS elevation is a good prognostic and predictive marker in all non-muscle invasive bladder cancer cases and in the subgroup receiving BCG. Elevation of FOXP3-TLS, TIM-3-TLS, and TIM-3/TLS is associated with longer RFS in patients receiving BCG. Combined TIM-3 and FOXP3 elevation is indicative of a low recurrence rate in NMIBC.
Collapse
Affiliation(s)
| | - Fatih Yılmaz
- Mardin Training and Research Hospital, Pathology Laboratory, Mardin, Turkey.
| |
Collapse
|