1
|
Sun L, Liu Y, Sun Q, Wang G, Du B, Liu B, Gao T, Zhao P, Yang Y, Rong R. Polysaccharides from traditional Chinese medicine and their nano-formulated delivery systems for cancer immunotherapy. Carbohydr Polym 2025; 357:123416. [PMID: 40158963 DOI: 10.1016/j.carbpol.2025.123416] [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: 11/25/2024] [Revised: 02/11/2025] [Accepted: 02/17/2025] [Indexed: 04/02/2025]
Abstract
Cancer immunotherapy has evolved into a new generation strategy in the field of anti-tumor treatment. Polysaccharides derived from Traditional Chinese Medicine (TCM) are gaining recognition as powerful immunomodulators in cancer therapy, noted for their multi-target and multi-pathway actions. Owing to their beneficial properties such as water solubility, biocompatibility, and chemical structure modifiability, TCM polysaccharides can also serve as carriers for hydrophobic drugs in the development of innovative drug delivery systems, enhancing synergistic antitumor effects. In this article, we summarize the diverse mechanisms of immunoregulation by TCM polysaccharides in tumor therapy. The applications of these polysaccharides as both active ingredients and drug carriers within nanodelivery systems for cancer immunotherapy are also introduced. Additionally, extensive research on TCM polysaccharides in clinical settings has been collected. Furthermore, discussions are presented on the development prospects and challenges faced by these polysaccharides in the field of tumor immunotherapy. Our goal is to improve researchers' comprehension of TCM polysaccharides in cancer immunotherapy, providing promising strategies to optimize cancer treatment and benefit diverse patient populations.
Collapse
Affiliation(s)
- Linlin Sun
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Yuting Liu
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Qihui Sun
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Guimei Wang
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Baoxiang Du
- Qingdao Academy of Chinese Medical Sciences, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Bodong Liu
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Tian Gao
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Pan Zhao
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Yong Yang
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China; Collaborative Innovation Center for Antiviral Traditional Chinese Medicine in Shandong Province, Jinan 250355, PR China; Shandong Antiviral Engineering Research Center of Traditional Chinese Medicine, Jinan 250355, PR China.
| | - Rong Rong
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China.
| |
Collapse
|
2
|
Ji Y, Liu L, Liu Y, Ma Y, Ji Z, Wu X, Zhu Q. Exploring gene biomarkers and targeted drugs for ferroptosis and cuproptosis in osteosarcoma: A bioinformatic approach. ENVIRONMENTAL TOXICOLOGY 2025; 40:891-901. [PMID: 38546286 PMCID: PMC12069744 DOI: 10.1002/tox.24250] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/05/2024] [Accepted: 03/14/2024] [Indexed: 05/14/2025]
Abstract
Osteosarcoma predominantly affects adolescents and young adults and is characterized as a malignant bone tumor. In recent decades, substantial advancements have been achieved in both diagnosing and treating osteosarcoma. Resulting in enhanced survival rates. Despite these advancements, the intricate relationship between ferroptosis and cuproptosis genes in osteosarcoma remains inadequately understood. Leveraging TARGET and GEO datasets, we conducted Cox regression analysis to select prognostic genes from a cohort of 71 candidates. Subsequently, a novel prognostic model was engineered using the LASSO algorithm. Kaplan-Meier analysis demonstrated that patients stratified as low risk had a substantially better prognosis compared with their high-risk counterparts. The model's validity was corroborated by the area under the receiver operating characteristic (ROC) curve. Additionally, we ascertained independent prognostic indicators, including clinical presentation, metastatic status, and risk scores, and crafted a clinical scoring system via nomograms. The tumor immune microenvironment was appraised through ESTIMATE, CIBERSORT, and single-sample gene set enrichment analysis. Gene expression within the model was authenticated through PCR validation. The prognostic model, refined by Cox regression and the LASSO algorithm, comprised two risk genes. Kaplan-Meier curves confirmed a significantly improved prognosis for the low-risk group in contrast to those identified as high-risk. For the training set, the ROC area under the curve (AUC) values stood at 0.636, 0.695, and 0.729 for the 1-, 3-, and 5-year checkpoints, respectively. Although validation set AUCs were 0.738, 0.668, and 0.596, respectively. Immune microenvironmental analysis indicated potential immune deficiencies in high-risk patients. Additionally, sensitivity to three small molecule drugs was investigated in the high-risk cohort, informing potential immunotherapeutic strategies for osteosarcoma. PCR analysis showed increased mRNA levels of the genes FDX1 and SQLE in osteosarcoma tissues. This study elucidates the interaction of ferroptosis and cuproptosis genes in osteosarcoma and paves the way for more targeted immunotherapy.
Collapse
Affiliation(s)
- Yingnan Ji
- Central Hospital Affiliated to Shenyang Medical CollegeShenyangChina
| | - Lv Liu
- Benxi Central HospitalBenxiChina
| | - Yu Liu
- Central Hospital Affiliated to Shenyang Medical CollegeShenyangChina
| | - Yudong Ma
- Central Hospital Affiliated to Shenyang Medical CollegeShenyangChina
| | - Zhenhua Ji
- Central Hospital Affiliated to Shenyang Medical CollegeShenyangChina
| | - Xiaodan Wu
- Central Hospital Affiliated to Shenyang Medical CollegeShenyangChina
| | - Qi Zhu
- Central Hospital Affiliated to Shenyang Medical CollegeShenyangChina
| |
Collapse
|
3
|
Backman EA, Gardberg M, Luntamo L, Peurla M, Vahlberg T, Borghammer P, Stefanova N, Wenning G, Kaasinen V. Nigral Neuroinflammation and Dopaminergic Neurons in Parkinson's Disease and Atypical Parkinsonisms. Ann Neurol 2025; 97:1096-1109. [PMID: 39918108 DOI: 10.1002/ana.27202] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 01/14/2025] [Accepted: 01/16/2025] [Indexed: 05/17/2025]
Abstract
OBJECTIVE To investigate the role of neuroinflammation in the substantia nigra pars compacta (SNc) across different parkinsonian disorders-Parkinson's disease (PD), progressive supranuclear palsy (PSP), and multiple system atrophy (MSA)-by examining SNc dopaminergic neuron counts, neuroinflammatory T cells, and microglial activity. METHODS Postmortem neuropathological samples were collected from 79 individuals (PD, n = 38; PSP, n = 15; MSA, n = 14; controls, n = 12). The density of SNc tyrosine hydroxylase (TH)-positive neurons, T cells (CD3+, CD4+, and CD8+), and Iba1 expression (Iba1-positive microglia/macrophages) were examined in the SNc and crus cerebri. Demographic and clinical data were gathered from patient histories. RESULTS PSP patients had 89 to 212% more nigral CD3+, CD4+, and CD8+ T cells compared to MSA patients (p < 0.04), 125 to 178% more CD3+ and CD4+ T cells than healthy controls (p < 0.002), and 95% more CD4+ T cells than PD patients (p = 0.001). Iba1 expression in the SNc was higher in PD patients than in MSA patients (p = 0.004), with no significant differences observed across other conditions. There was a negative association between disease duration and SNc CD3+ T cell density (p = 0.002), and a positive correlation between nigral dopaminergic neuron density and CD3+ density, CD8+ density, and Iba1 expression in PD patients. INTERPRETATION The study reveals distinctive neuroinflammatory patterns in the SNc, with T cell-mediated inflammation prominent in PSP and microglia-mediated inflammation in PD. PSP and MSA show greater SNc dopaminergic neuron loss compared to PD. Increased neuroinflammatory response is seen in earlier disease stages, diminishing with greater neuron loss, which may inform disease progression understanding and therapeutic strategies. ANN NEUROL 2025;97:1096-1109.
Collapse
Affiliation(s)
- Emmilotta A Backman
- Clinical Neurosciences, University of Turku, Turku, Finland
- Neurocenter, Turku University Hospital, Turku, Finland
| | - Maria Gardberg
- Tyks Laboratories, Department of Pathology, Turku University Hospital and Institute of Biomedicine, University of Turku, Turku, Finland
| | - Laura Luntamo
- Clinical Neurosciences, University of Turku, Turku, Finland
- Neurocenter, Turku University Hospital, Turku, Finland
| | - Markus Peurla
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Tero Vahlberg
- Department of Biostatistics, University of Turku and Turku University Hospital, Turku, Finland
| | - Per Borghammer
- Department of Nuclear Medicine and PET, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Nadia Stefanova
- Division of Neurobiology, Department of Neurology, Medical University Innsbruck, Innsbruck, Austria
| | - Gregor Wenning
- Division of Neurobiology, Department of Neurology, Medical University Innsbruck, Innsbruck, Austria
| | - Valtteri Kaasinen
- Clinical Neurosciences, University of Turku, Turku, Finland
- Neurocenter, Turku University Hospital, Turku, Finland
| |
Collapse
|
4
|
Yu Y, Jin C, Fu R, Han L, Fu B, Li Q, Cheng Y, Leng J. Splenic comparative transcriptome analysis reveals the immunological mode of undomesticated Gayal (Bos frontalis) for adapting to harsh environments. BMC Genomics 2025; 26:514. [PMID: 40394466 DOI: 10.1186/s12864-025-11718-3] [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: 11/12/2024] [Accepted: 05/15/2025] [Indexed: 05/22/2025] Open
Abstract
BACKGROUND The utilization of transcriptome technology in the identification of pivotal regulatory genes associated with immunity is of paramount importance. Previous studies have shown that undomesticated gayal (Bos frontalis) may have higher humoral responses which is comparable to yaks. However, research on immune function of gayal is limited, and comparisons with different breeds are rarely reported. The objective of this study was to inspect the immune status and compare splenic differential expression genes (DEGs) through comparative transcriptome analysis of gayal and domesticated local cattle (Yunan yellow cattle). RESULTS Serum immunological status investigation showed the better humoral immune status and lower levels of pro-inflammatory cytokines of gayal when compared to the local cattle. Spleen RNA-seq showed that 708 DEGs (365 up- and 343 down-regulated genes) were obtained between the gayal and local cattle. Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) enrichment analysis showed that immune system pathways, immune disease pathways, and chemotaxis-related molecular function of gayal were significantly enriched, whereas T cell-related cellular component and biological process were downregulated in the gayal. Correlation analysis shown that CD1, CD36, CD38, CD179a, CD179b, CXCL8, IGCGAMMA, IGH, IGHG1, IGLL1, IL1R2, SERPINB, and SERPINB4 had positive correlations with splenic IgA, IgD, IgE, IgG, and IgM, respectively (R > 0.5, P < 0.05). ANPEP, BVD1.23, CD1E, CD3D, CD3E, CD3G, CD5, CD8 A, HBB, IDO1, LCK, MGC126945, MHC1, TRAV, TRBV, and ZAP70 had negative correlations with splenic IgA, IgD, IgE, IgG, and IgM, respectively (R < -0.5, P < 0.05). CONCLUSIONS Our results reveal the immunological mode of gayal with high-level humoral immunity and enhanced splenic immunoglobulin gene expression and B cell differentiation, which may enable gayal to adapt to the harsh environments.
Collapse
Affiliation(s)
- Ye Yu
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
- Key Laboratory of Animal Nutrition and Feed Science of Yunnan Province, Yunnan Agricultural University, Kunming, 650201, China
| | - Chunjia Jin
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
- Key Laboratory of Animal Nutrition and Feed Science of Yunnan Province, Yunnan Agricultural University, Kunming, 650201, China
| | - Runqi Fu
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
- Key Laboratory of Animal Nutrition and Feed Science of Yunnan Province, Yunnan Agricultural University, Kunming, 650201, China
| | - Lin Han
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
- Key Laboratory of Animal Nutrition and Feed Science of Yunnan Province, Yunnan Agricultural University, Kunming, 650201, China
| | - Binlong Fu
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
- Key Laboratory of Animal Nutrition and Feed Science of Yunnan Province, Yunnan Agricultural University, Kunming, 650201, China
| | - Qian Li
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
- Key Laboratory of Animal Nutrition and Feed Science of Yunnan Province, Yunnan Agricultural University, Kunming, 650201, China
| | - Yanfen Cheng
- Laboratory of Gastrointestinal Microbiology, National Center for International Research On Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, 210095, China
| | - Jing Leng
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China.
- Key Laboratory of Animal Nutrition and Feed Science of Yunnan Province, Yunnan Agricultural University, Kunming, 650201, China.
| |
Collapse
|
5
|
Aviles-Huerta D, Del Pizzo R, Kowar A, Baig AH, Palazzo G, Stepanova E, Amaya Ramirez CC, D'Agostino S, Ratto E, Pechincha C, Siefert N, Engel H, Du S, Cadenas-De Miguel S, Miao B, Cruz-Vilchez VM, Müller-Decker K, Elia I, Sun C, Palm W, Loayza-Puch F. Dual Ribosome Profiling reveals metabolic limitations of cancer and stromal cells in the tumor microenvironment. Nat Commun 2025; 16:4652. [PMID: 40389477 DOI: 10.1038/s41467-025-59986-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 05/09/2025] [Indexed: 05/21/2025] Open
Abstract
The tumor microenvironment (TME) influences cancer cell metabolism and survival. However, how immune and stromal cells respond to metabolic stress in vivo, and how nutrient limitations affect therapy, remains poorly understood. Here, we introduce Dual Ribosome Profiling (DualRP) to simultaneously monitor translation and ribosome stalling in multiple tumor cell populations. DualRP reveals that cancer-fibroblast interactions trigger an inflammatory program that reduces amino acid shortages during glucose starvation. In immunocompetent mice, we show that serine and glycine are essential for optimal T cell function and that their deficiency impairs T cell fitness. Importantly, immune checkpoint blockade therapy imposes amino acid restrictions specifically in T cells, demonstrating that therapies create distinct metabolic demands across TME cell types. By mapping codon-resolved ribosome stalling in a cell‑type‑specific manner, DualRP uncovers metabolic crosstalk that shapes translational programs. DualRP thus offers a powerful, innovative approach for dissecting tumor cell metabolic interplay and guiding combined metabolic-immunotherapeutic strategies.
Collapse
Affiliation(s)
- Daniela Aviles-Huerta
- Translational Control and Metabolism, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg, Germany
- Faculty of Biosciences, University of Heidelberg, Heidelberg, Germany
| | - Rossella Del Pizzo
- Translational Control and Metabolism, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg, Germany
- Faculty of Biosciences, University of Heidelberg, Heidelberg, Germany
| | - Alexander Kowar
- Translational Control and Metabolism, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg, Germany
- Faculty of Biosciences, University of Heidelberg, Heidelberg, Germany
| | - Ali Hyder Baig
- Translational Control and Metabolism, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg, Germany
| | - Giuliana Palazzo
- Translational Control and Metabolism, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg, Germany
- Faculty of Biosciences, University of Heidelberg, Heidelberg, Germany
| | - Ekaterina Stepanova
- Translational Control and Metabolism, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg, Germany
| | - Cinthia Claudia Amaya Ramirez
- Translational Control and Metabolism, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg, Germany
| | - Sara D'Agostino
- Translational Control and Metabolism, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg, Germany
| | - Edoardo Ratto
- Faculty of Biosciences, University of Heidelberg, Heidelberg, Germany
- Division of Cell Signaling and Metabolism, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg, Germany
| | - Catarina Pechincha
- Faculty of Biosciences, University of Heidelberg, Heidelberg, Germany
- Division of Cell Signaling and Metabolism, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg, Germany
| | - Nora Siefert
- Faculty of Biosciences, University of Heidelberg, Heidelberg, Germany
- Division of Cell Signaling and Metabolism, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg, Germany
| | - Helena Engel
- Faculty of Biosciences, University of Heidelberg, Heidelberg, Germany
- Immune Regulation in Cancer, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg, Germany
| | - Shangce Du
- Immune Regulation in Cancer, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg, Germany
- Faculty of Medicine, University of Heidelberg, Heidelberg, Germany
| | | | - Beiping Miao
- Immune Regulation in Cancer, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg, Germany
| | - Victor M Cruz-Vilchez
- Translational Control and Metabolism, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg, Germany
| | - Karin Müller-Decker
- Core Facility Tumor Models, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg, Germany
| | - Ilaria Elia
- Department of Cellular and Molecular Medicine, KU Leuven, 3000, Leuven, Belgium
| | - Chong Sun
- Immune Regulation in Cancer, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg, Germany
| | - Wilhelm Palm
- Division of Cell Signaling and Metabolism, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg, Germany
| | - Fabricio Loayza-Puch
- Translational Control and Metabolism, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, Heidelberg, Germany.
| |
Collapse
|
6
|
Zhang X, Wang Y, E Q, Naveed M, Wang X, Liu Y, Li M. The biological activity and potential of probiotics-derived extracellular vesicles as postbiotics in modulating microbiota-host communication. J Nanobiotechnology 2025; 23:349. [PMID: 40380331 PMCID: PMC12082936 DOI: 10.1186/s12951-025-03435-6] [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: 05/01/2025] [Indexed: 05/19/2025] Open
Abstract
Probiotics such as Lactobacillus and Bifidobacterium spp. have been shown to be critical for maintaining host homeostasis. In recent years, key compounds of postbiotics derived from probiotic metabolism and cellular secretion have been identified for their role in maintaining organ immunity and regulating intestinal inflammation. In particular, probiotic-derived extracellular vesicles (PEVs) can act as postbiotics, maintaining almost the same functional activity as probiotics. They also have strong biocompatibility and loading capacity to carry exogenous or parental active molecules to reach distal organs to play their roles. This provides a new direction for understanding the intrinsic microbiota-host communication mechanism. However, most current studies on PEVs are limited to their functional effects/benefits, and their specific physicochemical properties, composition, intrinsic mechanisms for maintaining host homeostasis, and possible threats remain to be explored. Here, we review and summarize the unique physicochemical properties of PEVs and their bioactivities and mechanisms in mediating microbiota-host communication, and elucidate the limitations of the current research on PEVs and their potential application as postbiotics.
Collapse
Affiliation(s)
- Xiaoming Zhang
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Ye Wang
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Qiyu E
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Muhammad Naveed
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Xiuli Wang
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Yinhui Liu
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Ming Li
- College of Basic Medical Science, Dalian Medical University, Dalian, China.
| |
Collapse
|
7
|
Dai W, Li Y, Wu S, Wang Q, Zheng X, Zhang J, Han X, Zhou Y. Identification of MAGE-A10 specific T cell receptor promising in immunotherapy of hepatocellular carcinoma. Int J Biol Macromol 2025:144243. [PMID: 40379175 DOI: 10.1016/j.ijbiomac.2025.144243] [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: 03/20/2025] [Revised: 05/12/2025] [Accepted: 05/13/2025] [Indexed: 05/19/2025]
Abstract
Due to the limitations of current treatment strategies, hepatocellular carcinoma (HCC) continues to impose a severe burden on people's health. In the process of exploring novel therapies, T cell receptor-engineered T cell (TCR-T) therapy has been extensively developed in HCC immunotherapy. Melanoma-associated antigen family A member 10 (MAGE-A10) is a cancer-testis antigen (CTA), specifically expressed on HCC cells. However, the identification of TCR-T targeting MAGE-A10 in HCC remains rarely discussed. In this study, single-cell RNA sequencing (scRNA-seq) and TCR sequencing (scTCR-seq) were performed on samples from HCC patients. The cellular landscape of HCC was illustrated through a single-cell atlas. Reactive T cells were defined based on the matched T cells. Additionally, most reactive T cells were enriched in CD4_CD69_Th, CD4_FOXP3_Treg, CD4_CXCL13_TEX, and CD8_CXCL13_TEX. GLIPH2 was utilized to cluster TCRs from reactive T cells, enabling the identification of reactive TCRs. TCRMatch predicted MAGE-A10 as a specific antigen recognized by one of the reactive TCRs. Furthermore, the affinity assessments between human leukocyte antigen (HLA), epitope of MAGE-A10, and the identified TCR were performed with NetMHCpan and DLpTCR. Finally, cytotoxicity assays indicated the specific recognition and killing of MAGE-A10-TCR-T cells against HCC cells, paving the way for TCR-T immunotherapy in HCC.
Collapse
Affiliation(s)
- Wei Dai
- Tumor Biological Diagnosis and Treatment Center, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou 213003, China; Institute of Cell Therapy, Soochow University, Changzhou 213003, China
| | - Yuanqi Li
- Tumor Biological Diagnosis and Treatment Center, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou 213003, China; Institute of Cell Therapy, Soochow University, Changzhou 213003, China
| | - Shaoxian Wu
- Tumor Biological Diagnosis and Treatment Center, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou 213003, China; Institute of Cell Therapy, Soochow University, Changzhou 213003, China
| | - Qi Wang
- Tumor Biological Diagnosis and Treatment Center, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou 213003, China; Institute of Cell Therapy, Soochow University, Changzhou 213003, China
| | - Xiao Zheng
- Tumor Biological Diagnosis and Treatment Center, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou 213003, China; Institute of Cell Therapy, Soochow University, Changzhou 213003, China
| | - Jinping Zhang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Xiao Han
- Institute of Pediatrics, Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou 510623, China.
| | - You Zhou
- Tumor Biological Diagnosis and Treatment Center, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou 213003, China; Institute of Cell Therapy, Soochow University, Changzhou 213003, China.
| |
Collapse
|
8
|
Zhang K, Zhang Y, Xiang P, Wang Y, Li Y, Jiang S, Zhang Y, Chen M, Su W, Li X, Li S. Advances in T Cell-Based Cancer Immunotherapy: From Fundamental Mechanisms to Clinical Prospects. Mol Pharm 2025. [PMID: 40359327 DOI: 10.1021/acs.molpharmaceut.4c01502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
T cells and their T cell receptors (TCRs) play crucial roles in the adaptive immune system's response against pathogens and tumors. However, immunosenescence, characterized by declining T cell function and quantity with age, significantly impairs antitumor immunity. Recent years have witnessed remarkable progress in T cell-based cancer treatments, driven by a deeper understanding of T cell biology and innovative screening technologies. This review comprehensively examines T cell maturation mechanisms, T cell-mediated antitumor responses, and the implications of thymic involution on T cell diversity and cancer prognosis. We discuss recent advances in adoptive T cell therapies, including tumor-infiltrating lymphocyte (TIL) therapy, engineered T cell receptor (TCR-T) therapy, and chimeric antigen receptor T cell (CAR-T) therapy. Notably, we highlight emerging DNA-encoded library technologies in mammalian cells for high-throughput screening of TCR-antigen interactions, which are revolutionizing the discovery of novel tumor antigens and optimization of TCR affinity. The review also explores strategies to overcome challenges in the solid tumor microenvironment and emerging approaches to enhance the efficacy of T cell therapy. As our understanding of T cell biology deepens and screening technologies advances, T cell-based immunotherapies show increasing promise for delivering durable clinical benefits to a broader patient population.
Collapse
Affiliation(s)
- Kaili Zhang
- Department of Molecular Pharmacology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Yi Zhang
- Department of Molecular Pharmacology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Pan Xiang
- Department of Molecular Pharmacology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Yi Wang
- Department of Molecular Pharmacology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Yifan Li
- Department of Molecular Pharmacology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Shuze Jiang
- Department of Molecular Pharmacology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Yuxuan Zhang
- Department of Molecular Pharmacology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Min Chen
- Department of Molecular Pharmacology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Weijun Su
- School of Medicine, Nankai University, Tianjin 300071, China
| | - Xiaoling Li
- Cell Biotechnology Laboratory, Tianjin Cancer Hospital Airport Hospital, Tianjin 300308, China
- National Clinical Research Center for Cancer, Tianjin 300060, China
- Haihe Laboratory of Synthetic Biology, Tianjin 300090, China
| | - Shuai Li
- Department of Molecular Pharmacology, Tianjin Medical University Cancer Institute and Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| |
Collapse
|
9
|
Zou Y, Li S, Li Y, Zhang D, Zheng M, Shi B. Glioblastoma Cell Derived Exosomes as a Potent Vaccine Platform Targeting Primary Brain Cancers and Brain Metastases. ACS NANO 2025; 19:17309-17322. [PMID: 40312770 DOI: 10.1021/acsnano.4c14573] [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: 05/03/2025]
Abstract
Glioblastoma multiforme (GBM) is the most prevalent brain tumor that remains incurable up to now. The rapid advancement of immunotherapy makes vaccines a promising therapeutic approach for GBM. However, current vaccine platforms, such as peptides, dendritic cells, mRNA, and viral vectors, are subject to limitations such as inadequate antigen loading, insufficient immune system activation, ineffective vector delivery, complicated fabrication process, and complex formulation. Here, we developed a GBM tumor cell derived homologous exosomal nanovaccine that does not need to carry any additional tumor antigens and leads to the activation of antigen-presenting cells (APCs) in lymph nodes, increasing the proportion of immune cells (matured dendritic cells, cytotoxic T cells, and memory T cells) and in turn promoting the expression of cytokines (TNF-α, IL-6, and IFN-γ), which effectively stimulates innate immunity to trigger durable protective immunity against tumor cell insult. Our nanovaccine platform possesses efficient dual-targeting capability to lymph nodes and the brain. More importantly, the developed exosomal nanovaccines protected 100% of treated mice by inducing sustained and strong immunity against GL261-luc GBM tumor cells, resulting in 100% mouse survival (8/8) up to 5 months. Our nanovaccines also induced antitumor immune responses in the immunosuppressed CT2A-luc GBM mouse model with greatly improved survival compared to control mice. Exosomal nanovaccines also demonstrated effectiveness in preventing brain metastasis in the B16F10-luc melanoma malignant brain metastasis mouse model, and the mice showed notably improved survival rates. Our simple and potent exosomes offer a versatile platform for clinical translation as individualized vaccine therapy.
Collapse
Affiliation(s)
- Yan Zou
- The Zhongzhou Laboratory for Integrative Biology, Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
- Macquarie Medical School, Faculty of Medicine, Human Health Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Shanshan Li
- The Zhongzhou Laboratory for Integrative Biology, Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Yundong Li
- The Zhongzhou Laboratory for Integrative Biology, Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Dongya Zhang
- The Zhongzhou Laboratory for Integrative Biology, Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Meng Zheng
- The Zhongzhou Laboratory for Integrative Biology, Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Bingyang Shi
- The Zhongzhou Laboratory for Integrative Biology, Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
- School of Biomedical Engineering, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| |
Collapse
|
10
|
Yosef O, Cohen-Daniel L, Shamriz O, Bar-On Z, Salaymeh W, Saragovi A, Abramovich I, Agranovich B, Lutz V, Tam J, Permyakova A, Gottlieb E, Huber M, Berger M. Metabolic reprogramming driven by Ant2 deficiency augments T Cell function and anti-tumor immunity in mice. Nat Commun 2025; 16:4292. [PMID: 40341170 PMCID: PMC12062294 DOI: 10.1038/s41467-025-59310-3] [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: 08/29/2024] [Accepted: 04/18/2025] [Indexed: 05/10/2025] Open
Abstract
T cell activation requires a substantial increase in NAD+ production, often exceeding the capacity of oxidative phosphorylation (OXPHOS). To investigate how T cells adapt to this metabolic challenge, we generate T cell-specific ADP/ATP translocase-2 knockout (Ant2-/-) mice. Loss of Ant2, a crucial protein mediating ADP/ATP exchange between mitochondria and cytoplasm, induces OXPHOS restriction by limiting ATP synthase activity, thereby impeding NAD+ regeneration. Interestingly, Ant2-/- naïve T cells exhibit enhanced activation, proliferation and effector functions compared to wild-type controls. Metabolic profiling reveals that these T cells adopt an activated-like metabolic program with increased mitobiogenesis and anabolism. Lastly, pharmacological inhibition of ANT in wild-type T cells recapitulates the Ant2-/- phenotype and improves adoptive T cell therapy of cancer in mouse models. Our findings thus suggest that Ant2-deficient T cells bypass the typical metabolic reprogramming required for activation, leading to enhanced T cell function and highlighting the therapeutic potential of targeting ANT for immune modulation.
Collapse
Affiliation(s)
- Omri Yosef
- The Institute for Medical Research Israel-Canada (IMRIC), Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Leonor Cohen-Daniel
- The Institute for Medical Research Israel-Canada (IMRIC), Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Oded Shamriz
- The Institute for Medical Research Israel-Canada (IMRIC), Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Zahala Bar-On
- The Institute for Medical Research Israel-Canada (IMRIC), Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Wajeeh Salaymeh
- The Institute for Medical Research Israel-Canada (IMRIC), Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Amijai Saragovi
- The Institute for Medical Research Israel-Canada (IMRIC), Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ifat Abramovich
- Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Bella Agranovich
- Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Veronika Lutz
- Institute of Systems Immunology, Philipps University of Marburg, Marburg, Germany
| | - Joseph Tam
- Obesity and Metabolism Laboratory, The Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Anna Permyakova
- Obesity and Metabolism Laboratory, The Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Eyal Gottlieb
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Magdalena Huber
- Institute of Systems Immunology, Philipps University of Marburg, Marburg, Germany
| | - Michael Berger
- The Institute for Medical Research Israel-Canada (IMRIC), Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
11
|
Martínez Villarreal A, Gantchev J, Xie P, Lefrançois P, Ramchatesingh B, Litvinov IV. Memory T-Cell Phenotype in Cutaneous T-Cell Lymphoma Is Modified by Germline Gene Gametocyte Specific Factor 1. Exp Dermatol 2025; 34:e70123. [PMID: 40369846 PMCID: PMC12078864 DOI: 10.1111/exd.70123] [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: 10/06/2024] [Revised: 04/10/2025] [Accepted: 05/01/2025] [Indexed: 05/16/2025]
Abstract
Cutaneous T-cell lymphoma (CTCL) is a heterogeneous group of lymphoproliferative disorders characterised by skin infiltration by malignant memory T cells. While most patients will present with an indolent disease, others will follow a highly aggressive clinical course. Currently, defining disease prognosis remains challenging. Ectopic expression of gametocyte-specific factor 1 (GTSF1) has emerged as a potential prognostic biomarker. However, its contribution to CTCL carcinogenesis remains unknown. Here, we report that GTSF1 contributes to carcinogenesis by partially modifying the memory/effector phenotype of the malignant T cells. GTSF1 knockdown in CTCL cells led to T-cell activation and production of IFNγ and TNFα. Advanced stages of the disease are associated with decreased production of these cytokines. Notably, we show that patients classified with high expression of GTSF1 are associated with a worse disease prognosis. Taken together, our findings indicate that GTSF1 expression in CTCL cells allows them to acquire memory T-cell phenotype. Malignant memory T cells have a decreased production of immune-responsive cytokines, leading to a diminished immune response and disease progression. GTSF1 is an important candidate as a prognostic biomarker. Furthermore, understanding the specific function of GTSF1 might help develop novel targeted treatment options for CTCL patients.
Collapse
Affiliation(s)
- Amelia Martínez Villarreal
- Faculty of Medicine and Health Sciences, Research Institute of the McGill University Health CentreMcGill UniversityMontrealQuebecCanada
- Division of Experimental Medicine, Faculty of Medicine and Health SciencesMcGill UniversityMontrealQuebecCanada
| | - Jennifer Gantchev
- Department of NeurosurgeryBrigham and Women's Hospital, Harvard Medical SchoolBostonMassachusettsUSA
| | - Pingxing Xie
- Division of Dermatology, Faculty of Medicine and Health SciencesMcGill UniversityMontrealQuebecCanada
| | - Philippe Lefrançois
- Division of Experimental Medicine, Faculty of Medicine and Health SciencesMcGill UniversityMontrealQuebecCanada
- Division of Dermatology, Faculty of Medicine and Health SciencesMcGill UniversityMontrealQuebecCanada
- Lady Davis Institute for Medical ResearchJewish General Hospital, McGill UniversityMontrealQuebecCanada
| | - Brandon Ramchatesingh
- Faculty of Medicine and Health Sciences, Research Institute of the McGill University Health CentreMcGill UniversityMontrealQuebecCanada
- Division of Experimental Medicine, Faculty of Medicine and Health SciencesMcGill UniversityMontrealQuebecCanada
| | - Ivan V. Litvinov
- Faculty of Medicine and Health Sciences, Research Institute of the McGill University Health CentreMcGill UniversityMontrealQuebecCanada
- Division of Experimental Medicine, Faculty of Medicine and Health SciencesMcGill UniversityMontrealQuebecCanada
- Division of Dermatology, Faculty of Medicine and Health SciencesMcGill UniversityMontrealQuebecCanada
| |
Collapse
|
12
|
Zheng J, Liu W, Wang X, Li H, Wang Z, Ai Z. Curcumin enhances anti-tumor immunity in anaplastic thyroid carcinoma by elevating CD8+ T cell function and downregulating the AKT/mTORC1/STAT3/PD-L1 axis. Pathol Res Pract 2025; 269:155898. [PMID: 40101549 DOI: 10.1016/j.prp.2025.155898] [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: 11/01/2024] [Revised: 01/28/2025] [Accepted: 03/03/2025] [Indexed: 03/20/2025]
Abstract
Curcumin, a compound isolated from turmeric, has been found to have promising anti-tumor effects in various cancers, including anaplastic thyroid carcinoma (ATC). However, the molecular mechanism of curcumin in ATC remains largely unclear. CD8 +T cells could eliminate rapidly proliferating malignant cells, whereas interaction between programmed death-1 (PD-1) and programmed cell death ligand 1 (PD-L1) could inhibit the activation and functions of CD8 + T cells. Thus, we aimed to explore whether curcumin could inhibit ATC progression via regulating CD8 + T cells and PD-L1 expression. The protein expression of PD-L1 in ATC cells was detected by western blot assay. Additionally, a syngeneic mouse model was used to assess the effect of curcumin or/and anti-PD-1 treatment on tumorigenesis in vivo. The effect of curcumin on CD8 +T cell function was investigated by flow cytometry in vitro and in vivo. The results indicated curcumin notably suppressed ATC cell proliferation, migration and invasion and induced cell apoptosis. Additionally, curcumin could reduce PD-L1 level in ATC cells through inactivating AKT/mTORC1/STAT3 signaling. Meanwhile, curcumin obviously elevated CD8 + T cell function by elevating the number of IFN-γ producing CD8 + T cells. Furthermore, curcumin or anti-PD-L1 treatment could enhance anti-tumor immunity by increasing infiltration of CD8 + T cells in tumor tissues in vivo. As expected, compared to the single treatment, combination curcumin and anti-PD-1 treatment further elevated CD8 + T cell function in vivo, thereby potentiating anti-tumor immunity in ATC. Collectively, curcumin could enhance anti-tumor immunity in ATC by elevating CD8 + T cell function and inactivating the AKT/mTORC1/STAT3/PD-L1 axis. Our findings demonstrated a novel mechanism of the anti-tumor effects of curcumin in ATC.
Collapse
Affiliation(s)
- Jiaojiao Zheng
- Department of General Surgery (Thyroid & Breast), Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Wei Liu
- Department of General Surgery (Thyroid & Breast), Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xiaolong Wang
- Department of General Surgery, Shanghai Xuhui Central Hospital, Fudan University, Shanghai 200032, China
| | - He Li
- Department of General Surgery (Thyroid & Breast), Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Zhenglin Wang
- Department of General Surgery (Thyroid & Breast), Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Zhilong Ai
- Department of General Surgery (Thyroid & Breast), Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| |
Collapse
|
13
|
Ma T, Zhang T, Miao F, Liu J, Zhu Q, Chen Z, Tai Z, He Z. Alopecia Areata: Pathogenesis, Diagnosis, and Therapies. MedComm (Beijing) 2025; 6:e70182. [PMID: 40260013 PMCID: PMC12010142 DOI: 10.1002/mco2.70182] [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: 10/09/2024] [Revised: 03/17/2025] [Accepted: 03/26/2025] [Indexed: 04/23/2025] Open
Abstract
Alopecia areata (AA) is a complex, chronic inflammatory skin disorder characterized by unpredictable, nonscarring hair loss, affecting millions worldwide. Its pathogenesis remains poorly understood, driven by intricate interactions among immune dysregulation, genetic predisposition, and environmental triggers. Despite significant advances in identifying these contributing factors, substantial gaps persist in our understanding of the full spectrum of AA's molecular mechanisms and in the development of effective therapeutic approaches. This review aims to comprehensively explore the immunological, genetic, epigenetic, and environmental factors underlying AA, with a focus on immune-mediated mechanisms. We also evaluate diagnostic approaches and recent advancements in assessing disease severity. Furthermore, the review discusses evolving therapeutic options, including traditional therapies, biologics, small-molecule agents, and emerging treatments. The academic value of this work lies in its synthesis of current knowledge on the multifaceted nature of AA, providing insights for future research and clinical practice. By elucidating the interconnected factors underlying AA, this review seeks to advance both understanding and management of this prevalent, clinically challenging disorder.
Collapse
Affiliation(s)
- Tianyou Ma
- Department of PharmacyLonghua Hospital of Shanghai University of Traditional Chinese MedicineShanghaiChina
- Shanghai Skin Disease HospitalSchool of MedicineTongji UniversityShanghaiChina
- Shanghai Engineering Research Center of External Chinese MedicineShanghaiChina
| | - Tingrui Zhang
- Shanghai Skin Disease HospitalSchool of MedicineTongji UniversityShanghaiChina
- Shanghai Engineering Research Center of External Chinese MedicineShanghaiChina
| | - Fengze Miao
- Shanghai Skin Disease HospitalSchool of MedicineTongji UniversityShanghaiChina
- Shanghai Engineering Research Center of External Chinese MedicineShanghaiChina
| | - Jun Liu
- Shanghai Skin Disease HospitalSchool of MedicineTongji UniversityShanghaiChina
- Shanghai Engineering Research Center of External Chinese MedicineShanghaiChina
| | - Quangang Zhu
- Shanghai Skin Disease HospitalSchool of MedicineTongji UniversityShanghaiChina
- Shanghai Engineering Research Center of External Chinese MedicineShanghaiChina
| | - Zhongjian Chen
- Shanghai Skin Disease HospitalSchool of MedicineTongji UniversityShanghaiChina
- Shanghai Engineering Research Center of External Chinese MedicineShanghaiChina
| | - Zongguang Tai
- Shanghai Skin Disease HospitalSchool of MedicineTongji UniversityShanghaiChina
- Shanghai Engineering Research Center of External Chinese MedicineShanghaiChina
| | - Zhigao He
- Department of PharmacyLonghua Hospital of Shanghai University of Traditional Chinese MedicineShanghaiChina
| |
Collapse
|
14
|
Keefe JA, Wang J, Song J, Ni L, Wehrens XHT. Immune cells and arrhythmias. Cardiovasc Res 2025; 121:382-395. [PMID: 39937651 PMCID: PMC12038251 DOI: 10.1093/cvr/cvaf017] [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: 11/06/2024] [Revised: 12/12/2024] [Accepted: 01/07/2025] [Indexed: 02/14/2025] Open
Abstract
Cardiac arrhythmias are a significant cause of morbidity and mortality worldwide. Emerging evidence has demonstrated that resident and infiltrating cardiac immune cells play direct, mechanistic roles in arrhythmia onset and progression. In this review, we provide a comprehensive summary and expert commentary on the role of each immune cell subtype in the pathogenesis of atrial and ventricular arrhythmias.
Collapse
Affiliation(s)
- Joshua A Keefe
- Cardiovascular Research Institute, Baylor College of Medicine, BCM335, One Baylor Plaza, Houston, TX 77030, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jian Wang
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, #1095 Jiefang Avenue, Wuhan 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Jiangping Song
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Xi Cheng District, Beijing 100037, China
- Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 167A Beilishi Road, Xi Cheng District, Beijing 100037, China
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences, 1021 Dongmen Rd N, Luohu District, Shenzhen, Guangdong Province, 518001, China
| | - Li Ni
- Division of Cardiology, Department of Internal Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, #1095 Jiefang Avenue, Wuhan 430030, China
- Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Wuhan, China
| | - Xander H T Wehrens
- Cardiovascular Research Institute, Baylor College of Medicine, BCM335, One Baylor Plaza, Houston, TX 77030, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
- Center for Space Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
15
|
Moadab A, Khorramdelazad H, Javar MTA, Nejad MSM, Mirzaie S, Hatami S, Mahdavi N, Ghaffari S, Yazdian FA. Unmasking a Paradox: Roles of the PD-1/PD-L1 Axis in Alzheimer's Disease-Associated Neuroinflammation. J Neuroimmune Pharmacol 2025; 20:46. [PMID: 40285967 DOI: 10.1007/s11481-025-10206-5] [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: 01/24/2025] [Accepted: 04/16/2025] [Indexed: 04/29/2025]
Abstract
Alzheimer's disease (AD) represents the most prevalent form of dementia, characterized by progressive cognitive impairment and chronic neuroinflammation. Immune checkpoint inhibitors (ICIs), including anti-programmed cell death (PD)-1 and anti-PD-L1, signify a revolutionary advancement in cancer treatment by preventing T-cell exhaustion; however, their therapeutic application in AD presents a conundrum. Hypothesis: Recent preclinical studies indicate that PD-1 inhibition in AD mouse models induces an interferon-gamma (IFN-γ)-mediated response, leading to increased recruitment of monocyte-derived macrophages into the brain, enhanced clearance of amyloid-beta (Aβ) plaques, and improved cognitive performance. Nonetheless, this therapeutic effect is counterbalanced by the potential for exacerbated neuroinflammation, as PD-1/PD-L1 blockade may potentiate pro-inflammatory T helper (Th)1 and Th17 responses. In this review, we critically discuss the pertinent pro-inflammatory and neuroprotective facets of T cell biology in the pathogenesis of AD, emphasizing the potential for modulation of the PD-1/PD-L1 axis to influence both Aβ clearance and the dynamics of neuroinflammatory processes. In summary, we determine that ICIs are promising tools for reducing AD pathology and improving cognition. However, it is essential to refine treatment protocols and carefully select patients to optimize neuroprotective effects while adequately considering inflammatory risks.
Collapse
Affiliation(s)
- Ali Moadab
- Department of Internal Medicine, School of Medicine, Ali-Ibn Abi-Talib Hospital, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Student Research Committee, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Hossein Khorramdelazad
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
- Student Research Committee, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| | - Mohammad Taha Akbari Javar
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Student Research Committee, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mohammad Saber Mohammadian Nejad
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Student Research Committee, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Shahrzad Mirzaie
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Student Research Committee, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Sina Hatami
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Student Research Committee, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Nima Mahdavi
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Student Research Committee, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Saeed Ghaffari
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Student Research Committee, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Fatemeh Askari Yazdian
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Student Research Committee, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| |
Collapse
|
16
|
Wu CJ, Yan J, Sun LP, Ma LQ, Li L, Liu J, Zhang JQ, Ren Y, Bi W. Correlation Analysis of Human Immunological Indicators and Nosocomial Infections, Along With Evaluation Value for Prognosis. J Immunol Res 2025; 2025:5539590. [PMID: 40313971 PMCID: PMC12045693 DOI: 10.1155/jimr/5539590] [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: 10/17/2023] [Revised: 01/19/2025] [Accepted: 03/10/2025] [Indexed: 05/03/2025] Open
Abstract
Objective: This study aimed to analyze the relevant risk factors for nosocomial infection (NI) in patients who were admitted to an emergency department, explore the correlation between each influencing factor and the risk of NI, and evaluate the application value of immunological indicators on the patient prognosis, all of which can provide reference for clinical guidance. Methods: We prospectively enrolled 128 patients meeting the inclusion criteria who visited the emergency department of Dongzhimen Hospital, Beijing University of Chinese Medicine, from January 1 to December 31, 2019. Basic information and serum samples were collected from the patients, and flow cytometry was used. T lymphocyte subgroups, CD3+CD4+and CD3+CD8+, and natural killer (NK) cells were measured. Patients were divided into infection group and control group according to whether nosocomial infection occurred within 48 h of admission. Age, gender, type of disease, APACHE II score, Charlton score, T lymphocyte subtypes, and NK cell values were compared, and a logistic multivariate regression analysis was conducted. A multifactor regression analysis was performed on various risk factors. The nomogram website was used to draw a nomogram model of meaningful indicators, and the receiver-operating characteristic (ROC) curve was based on experimental results. Results: Logistics multivariate regression analysis showed the Charlton score and NK cell count were independent risk factors for nosocomial infection. Cell counts for subsets CD3+CD4+ and CD3+CD8+ were protective factors, and the OR value and 95% CI were 5.199 (1.933-13.983), 1.248 (1.055-1.475), 0.851 (0.790-0.916), and 0.832 (0.711-0.973), p < 0.05. respectively. Statistical significance was set at p < 0.05.The nomogram model suggested that the area under the curve for predicting the risk of nosocomial infection was 0.920 (0.872-0.967), p < 0.001. Conclusion: Patients with low CD3+CD4+ and CD3+CD8+ T lymphocyte or high NK cell count as well as high Charlton score are more likely to have nosocomial infection. Then, we speculate that the risk of nosocomial infection within 48 h is also high for patients with underlying diseases and immune function that is affected and suppressed on admission, regardless of whether infection occurs during hospitalization.
Collapse
Affiliation(s)
- Cai-jun Wu
- Department of Emergency, Beijing Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Institute of Sepsis, Beijing University of Chinese Medicine, Beijing, China
| | - Jun Yan
- Department of Emergency, Beijing Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Institute of Sepsis, Beijing University of Chinese Medicine, Beijing, China
| | - Li-ping Sun
- Department of Infectious Diseases, Miyun District Hospital, Beijing, China
- Peking University First Hospital-Miyun Hospital, Beijing, China
| | - Lin-qin Ma
- Department of Emergency, Beijing Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Institute of Sepsis, Beijing University of Chinese Medicine, Beijing, China
| | - Lan Li
- Department of Emergency, Beijing Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Institute of Sepsis, Beijing University of Chinese Medicine, Beijing, China
| | - Jin Liu
- Department of Emergency, Beijing Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Institute of Sepsis, Beijing University of Chinese Medicine, Beijing, China
| | - Jia-qi Zhang
- Department of Emergency, Beijing Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Institute of Sepsis, Beijing University of Chinese Medicine, Beijing, China
| | - Yang Ren
- Department of Emergency, Beijing Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Institute of Sepsis, Beijing University of Chinese Medicine, Beijing, China
| | - Wei Bi
- Department of Emergency, Beijing Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Institute of Sepsis, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
17
|
Gamal W, Mediavilla-Varela M, Kunta V, Sahakian E, Pinilla-Ibarz J. Impact of mitochondrial metabolism on T-cell dysfunction in chronic lymphocytic leukemia. Front Cell Dev Biol 2025; 13:1577081. [PMID: 40313718 PMCID: PMC12043688 DOI: 10.3389/fcell.2025.1577081] [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: 02/15/2025] [Accepted: 03/31/2025] [Indexed: 05/03/2025] Open
Abstract
T cells play a central role in anti-tumor immunity, yet their function is often compromised within the immunosuppressive tumor microenvironment, leading to cancer progression and resistance to immunotherapies. T-cell activation and differentiation require dynamic metabolic shifts, with mitochondrial metabolism playing a crucial role in sustaining their function. Research in cancer immunometabolism has revealed key mitochondrial abnormalities in tumor-infiltrating lymphocytes, including reduced mitochondrial capacity, depolarization, structural defects, and elevated reactive oxygen species. While these mitochondrial disruptions are well-characterized in solid tumors and linked to T-cell exhaustion, their impact on T-cell immunity in lymphoproliferative disorders remains underexplored. Chronic lymphocytic leukemia (CLL), the most prevalent chronic adult leukemia, is marked by profound T-cell dysfunction that limits the success of adoptive cell therapies. Emerging studies are shedding light on the role of mitochondrial disturbances in CLL-related T-cell dysfunction, but significant knowledge gaps remain. This review explores mitochondrial metabolism in T-cell exhaustion, emphasizing recent findings in CLL. We also discuss therapeutic strategies to restore T-cell mitochondrial function and identify key research gaps.
Collapse
Affiliation(s)
- Wael Gamal
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | - Melanie Mediavilla-Varela
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | - Vishaal Kunta
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | - Eva Sahakian
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | - Javier Pinilla-Ibarz
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| |
Collapse
|
18
|
Cao R, Jiao P, Zhang S, Li J, Liu Q. Predicting the Efficacy of Immune Checkpoint Inhibitors in Esophageal Cancer: Changes in Peripheral Blood Lymphocyte Subsets Before and After Immunotherapy. Cancer Manag Res 2025; 17:815-825. [PMID: 40256769 PMCID: PMC12009565 DOI: 10.2147/cmar.s503171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 04/04/2025] [Indexed: 04/22/2025] Open
Abstract
Background Immunotherapy has demonstrated potential in the treatment of esophageal cancer (EC); however, the overall response rate (ORR) remains below 30% among EC patients. Herein, the use of peripheral blood lymphocyte subsets as biomarkers was explored to evaluate the efficacy of immunotherapy in this patient population. Methods Sixty-three patients were enrolled. The patients were diagnosed with EC and treated with immune checkpoint inhibitors (ICIs) at The Fourth Hospital of Hebei Medical University from December 2019 to June 2023. Kaplan-Meier (KM) survival curves were used to reflect differences in survival benefit. The prognostic factors of survival were investigated using the Cox proportional hazards regression model for both univariate and multivariate analyses. Two-tailed P values were reported and statistical significance was defined as P < 0.05. Results The results of univariate and multifactorial Cox regression analysis for progression-free survival (PFS) revealed that only CD8+ T lymphocytes demonstrated a significant association with PFS (P = 0.034, P = 0.020). Additionally, the multifactorial Cox regression analysis results for overall survival (OS) revealed a significant association between natural killer (NK) cells and OS (P=0.049). Further, a systematic analysis was conducted on the CD8+ T cell biomarker. The KM survival curves indicated that the group with low CD8+ T cell levels experienced a significantly greater PFS benefit compared to the high CD8+ T cell group (P = 0.030). Conclusion The present study reveals that the reduction of both CD8+ T lymphocytes and NK cells in peripheral blood lymphocyte subsets after immunotherapy can serve as superior predictors for the effectiveness of ICIs in patients diagnosed with EC.
Collapse
Affiliation(s)
- Ruijie Cao
- Department of Immunology and Rheumatology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, People’s Republic of China
| | - Pengqing Jiao
- Department of Immunology and Rheumatology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, People’s Republic of China
| | - Shasha Zhang
- Department of Immunology and Rheumatology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, People’s Republic of China
| | - Jiasong Li
- Department of Immunology and Rheumatology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, People’s Republic of China
| | - Qingyi Liu
- Department of Cardiothoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, People’s Republic of China
| |
Collapse
|
19
|
Du X, Wei N, Wang A, Sun G. Liver cancer-specific prognostic model developed using endoplasmic reticulum stress-related LncRNAs and LINC01011 as a potential therapeutic target. BMC Med Genomics 2025; 18:71. [PMID: 40234922 PMCID: PMC12001585 DOI: 10.1186/s12920-025-02142-3] [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: 01/02/2025] [Accepted: 04/07/2025] [Indexed: 04/17/2025] Open
Abstract
Liver cancer is a serious malignancy worldwide, and long noncoding RNAs (lncRNAs) have been implicated in its prognosis.It remains unclear how lncRNAs related to endoplasmic reticulum stress (ERS) influence liver cancer prognosis. Here, we analyzed RNA and clinical data from the Cancer Genome Atlas and sourced ERS-related genes from the Molecular Signatures Database. Co-expression analysis identified ERS-related lncRNAs, and Cox regression analysis as well as least absolute shrinkage and selection operator regression highlighted three lncRNAs for a prognostic model. Based on median risk scores, we classified patients into two risk groups. The high-risk group displayed poor prognosis, and this finding was validated in the test set. According to consistency clustering, the patients were assigned to two clusters, and tumor microenvironment scores were computed. Patients with a high mutation burden had worse outcomes. Furthermore, immune infiltration analysis indicated more immune cells and mutations in checkpoint molecules among high-risk individuals. Drug sensitivity varied between the risk groups. LINC01011 was selected for functional assays. Colony formation assay and CCK-8 assay revealed that silencing LINC01011 suppressed liver cancer cell proliferation. Transwell and scratch assays indicated that silencing LINC01011 inhibited liver cancer cell migration. Western blotting assay revealed that inhibiting LINC01011 induced apoptosis and simultaneously inhibited epithelial-mesenchymal transition. These findings confirm the validity of the prognostic model and indicate that LINC01011 could serve as a potential research target.
Collapse
Affiliation(s)
- Xiao Du
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, 81 Meishan Road, Shushan District, Hefei, Anhui, 230000, China
| | - Ning Wei
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250000, China
- Department of Radiology, Division of Life Sciences and Medicine, the First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, 230000, China
| | - Anqi Wang
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, 81 Meishan Road, Shushan District, Hefei, Anhui, 230000, China
| | - Guoping Sun
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, 81 Meishan Road, Shushan District, Hefei, Anhui, 230000, China.
| |
Collapse
|
20
|
Yu J, Fu L, Wu R, Che L, Liu G, Ran Q, Xia Z, Liang X, Zhao G. Immunocytes in the tumor microenvironment: recent updates and interconnections. Front Immunol 2025; 16:1517959. [PMID: 40297580 PMCID: PMC12034658 DOI: 10.3389/fimmu.2025.1517959] [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: 10/27/2024] [Accepted: 03/11/2025] [Indexed: 04/30/2025] Open
Abstract
The tumor microenvironment (TME) is a complex, dynamic ecosystem where tumor cells interact with diverse immune and stromal cell types. This review provides an overview of the TME's evolving composition, emphasizing its transition from an early pro-inflammatory, immune-promoting state to a later immunosuppressive milieu characterized by metabolic reprogramming and hypoxia. It highlights the dual roles of key immunocytes-including T lymphocytes, natural killer cells, macrophages, dendritic cells, and myeloid-derived suppressor cells-which can either inhibit or support tumor progression based on their phenotypic polarization and local metabolic conditions. The article further elucidates mechanisms of immune cell plasticity, such as the M1/M2 macrophage switch and the balance between effector T cells and regulatory T cells, underscoring their impact on tumor growth and metastasis. Additionally, emerging therapeutic strategies, including checkpoint inhibitors and chimeric antigen receptor (CAR) T and NK cell therapies, as well as approaches targeting metabolic pathways, are discussed as promising avenues to reinvigorate antitumor immunity. By integrating recent molecular insights and clinical advancements, the review underscores the importance of deciphering the interplay between immunocytes and the TME to develop more effective cancer immunotherapies.
Collapse
Affiliation(s)
- Jiyao Yu
- Department of Ultrasound, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
- Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, United Kingdom
| | - Li Fu
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
- Department of Gastroenterology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Rui Wu
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
- Department of Neurosurgery, Jiangyou People’s Hospital, Mianyang, China
| | - Linyi Che
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Guodong Liu
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Qinwen Ran
- General Practice Department, Wufu Town Hospital, Chongqing, China
| | - Zhiwei Xia
- Department of Neurology, Hunan Aerospace Hospital, Hunan Normal University, Changsha, China
| | - Xisong Liang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Guanjian Zhao
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| |
Collapse
|
21
|
Yu Z, Xu Z, Fu T, Liu S, Cui J, Zhang B, Liang J, Pang C, Ke Y, Wang R, Tang Z, Gao Y, Du B, Feng Y, Zhao H, Xue G, Yan C, Gan L, Feng J, Fan Z, Yang Y, Huang L, Zhao S, Ying S, Gu Q, Yuan J. Parallel comparison of T cell and B cell subpopulations of adenoid hypertrophy and tonsil hypertrophy of children. Nat Commun 2025; 16:3516. [PMID: 40229254 PMCID: PMC11997228 DOI: 10.1038/s41467-025-58094-w] [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: 05/09/2024] [Accepted: 03/12/2025] [Indexed: 04/16/2025] Open
Abstract
The adenoids and tonsils are important immune organs of the nasopharynx that often become hypertrophic in childhood because of recurrent pathogen infection. However, the differences in the immune microenvironment of adenoid hypertrophy (AH) and tonsil hypertrophy (TH) are unclear. Here, we show the epidemiological characteristics and peripheral blood cell indices of 1209 pediatric patients (1-15 years old) diagnosed with AH, and find that AH is often accompanied by TH and characterized by specific changes in immune cell types. Single-cell RNA sequencing analysis show that 12 paired AH and TH samples contain large numbers of B, T cells and some exhausted effector memory CD4+ T cells. Compared with matched TH, AH have more naïve B cells and regulatory CD4+ T cells and less plasma B cells. Weaker antigen presentation and more significant immunosuppression are also observed in AH. In contrast, the number and cytotoxicity of cytotoxic CD8+ T cells decrease with AH grade. These findings will help our understanding of the immune response to nasopharyngeal infection.
Collapse
Affiliation(s)
- Zihui Yu
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Ziying Xu
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Tongtong Fu
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Shiyu Liu
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
- Military supplies and energy quality supervision station of Bejing, Beijing, 100071, China
| | - Jinghua Cui
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Bing Zhang
- Department of Otolaryngology, Capital Center For Children's Health, Capital Medical University, Beijing, 100020, China
| | - Jieqiong Liang
- Department of Otolaryngology, Capital Center For Children's Health, Capital Medical University, Beijing, 100020, China
| | - Chong Pang
- Department of Otolaryngology, Capital Center For Children's Health, Capital Medical University, Beijing, 100020, China
| | - Yuehua Ke
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Ruikun Wang
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
- Capital Institute of Pediatrics-Peking University Teaching Hospital, Beijing, 100020, China
| | - Zhijie Tang
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
- Capital Institute of Pediatrics-Peking University Teaching Hospital, Beijing, 100020, China
| | - Yagang Gao
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Bing Du
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Yanling Feng
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Hanqing Zhao
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Guanhua Xue
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Chao Yan
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Lin Gan
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Junxia Feng
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Zheng Fan
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Yang Yang
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Lijuan Huang
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Shuo Zhao
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China
| | - Sun Ying
- School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Qinglong Gu
- Department of Otolaryngology, Capital Center For Children's Health, Capital Medical University, Beijing, 100020, China.
| | - Jing Yuan
- Department of Bacteriology, Capital Institute of Pediatrics, Beijing, 100020, China.
| |
Collapse
|
22
|
Cai Y, Yang M, Liu X, Zhang L, Wang J, Sun Y. Effect of hydromorphone combined with ropivacaine caudal block on immune function after hypospadias surgery in children. BMC Anesthesiol 2025; 25:172. [PMID: 40211132 PMCID: PMC11987407 DOI: 10.1186/s12871-025-03053-7] [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: 10/29/2024] [Accepted: 04/02/2025] [Indexed: 04/12/2025] Open
Abstract
BACKGROUND This study aimed to evaluate the effects of caudal block anesthesia with hydromorphone-ropivacaine compared to ropivacaine alone on postoperative immune function and pain management in children undergoing hypospadias surgery. METHODS A total of 100 pediatric patients were randomly assigned to two groups: the Hydromorphone-Ropivacaine (HR) group and the Ropivacaine (R) group for caudal block anesthesia, with 50 patients in each group. The R group received 0.25% ropivacaine at a dose of 1 ml/kg, while the HR group received 0.25% ropivacaine (1 ml/kg) combined with hydromorphone (10 µg/kg). The maximum dose for both groups was capped at 30 ml (1 ml/kg). Anesthesia induction included intravenous administration of pentobarbital (0.01 mg/kg) and dexamethasone (0.15 mg/kg), followed by sevoflurane inhalation. All patients underwent ultrasound-guided caudal block anesthesia administered by the same anesthetist. Primary outcomes included the distribution of T lymphocyte subsets (CD3+, CD4+, CD8+, and CD4+/CD8 + ratios) measured at five time points: pre-anesthesia (T0), end of surgery (T1), 24 h postoperatively (T2), 48 h postoperatively (T3), and 72 h postoperatively (T4). Secondary outcomes included postoperative pain scores assessed using the Modified Children's Hospital of Eastern Ontario Pain Scale (M-CHEOPS) at 1, 6, 12, 18, and 24 h postoperatively, sedation levels evaluated using the Ramsay sedation scale at the same time points, and the incidence of postoperative adverse events. RESULTS The HR group exhibited significant reductions in CD3+, CD4+, and CD4+/CD8+ ratios at T1, T2, and T3 compared to baseline (T0) (p < 0.001). At all postoperative time points (T1, T2, T3, T4), the HR group demonstrated significantly higher levels of CD3+, CD4+, and CD4+/CD8+ ratios compared to the R group (p < 0.001). By T4 (72 h postoperatively), immune markers in the HR group had largely normalized to baseline levels, whereas those in the R group remained significantly lower (p < 0.001). Postoperative pain, assessed using the Modified Children's Hospital of Eastern Ontario Pain Scale (M-CHEOPS), was significantly lower in the HR group at 6, 12, and 18 h postoperatively compared to the R group (p < 0.001). The HR group also exhibited a longer duration of analgesia and required fewer doses of rescue analgesia within the first 24 h postoperatively (p = 0.046). Sedation levels, evaluated using the Ramsay sedation scale, showed significant differences between the groups at 1 h (p = 0.0087) and 6 h (p < 0.0001) postoperatively, with higher sedation scores observed in the HR group. There were no significant differences in heart rate, systolic blood pressure, diastolic blood pressure, mean arterial pressure, or oxygen saturation between the groups at any time point (p > 0.05). No significant differences were observed between the two groups in terms of postoperative adverse reactions (all p > 0.05). CONCLUSION Caudal block anesthesia with hydromorphone-ropivacaine offers enhanced postoperative pain relief and a lesser impact on immune function compared to ropivacaine alone in children undergoing hypospadias surgery. Further studies are warranted to explore the long-term effects on immune function.
Collapse
Affiliation(s)
- Yuzhu Cai
- Department of Anesthesiology, Anhui Provincial Children's Hospital, Hefei, 230051, China
| | - Mingwen Yang
- Department of Anesthesiology, Anhui Provincial Children's Hospital, Hefei, 230051, China
| | - Xinghui Liu
- Department of Anesthesiology, Anhui Provincial Children's Hospital, Hefei, 230051, China
| | - Lingli Zhang
- Department of Anesthesiology, Anhui Provincial Children's Hospital, Hefei, 230051, China
| | - Jun Wang
- Department of Anesthesiology, Anhui Provincial Children's Hospital, Hefei, 230051, China
| | - Yingying Sun
- Department of Anesthesiology, Anhui Provincial Children's Hospital, Hefei, 230051, China.
| |
Collapse
|
23
|
Li C, Xu D, Lu L, Peng S, Zhao H, Zeng C, Hu L, Guo X, Liu L, Huo F, Rong X, Geng Z, Lin P, Zhou X, Wang X, Hobeika A, Morse MA, Lyerly HK, Ren J. Clinical impact of concurrent autologous adoptive T cells immunotherapy in active COVID-19 infected cancer patients for chemotherapy. Infect Agent Cancer 2025; 20:23. [PMID: 40205403 PMCID: PMC11983847 DOI: 10.1186/s13027-025-00654-2] [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: 11/17/2024] [Accepted: 03/28/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND The concurrent presence of COVID-19 infection in advanced cancer patients has increased the mortality since the compromised immunity was inevitably worsen. The role and clinical impact of autologous adoptive T cell immunotherapy (ACT) designed for anti-cancer treatment were not known in such circumstances. The safety and potential immune reconstitution of concurrent ACT in advanced cancer patients with active COVID-19 infection have yet unknown as well. The effect of infused ACT on the symptom severity manifestation should be summarized. METHODS In this respectively clinical observation study, patients were non-randomized enrolled from the two centers according to the regular therapeutic plans including stage IV cancer patients for scheduled ACT, chemotherapy, cancer patients with symptomatic COVID-19 but without ACT, neither cancer or non-ACT but symptomatic cases of COVID-19 infection. We have incorporated the age-adjusted Charlson comorbidity index (aCCI) for each patient to compare the prognosis of the three groups. All patients were planned for the scheduled standard anti-cancer therapeutic considerations, chemotherapy plus ACT as planned as well as the supportive care.The clinical efficacy and impact of ACT on cancer patients within the 3 months from the peripheral blood apheresis, dendritic cell (DC) and cytokine induced killer T cell (CIK-T ) infusion and subsequent co-existence of COVID-19 infection were recorded as the primary objective. During the same period, the cancer cases without ACT and others were collected to compare the occurrence of both severe and death rate respectively. RESULTS There were 123 patients (35 of ACT, 23 of non-ACT, 65 of non-cancer) with similar aCCI. There were similar cohort-level COVID-19 in-hospital case fatality rates consistent with previously reported data for non-cancer (26.2%, 17/65) and non-ACT cancer (52.2%, 12/23) among those admitted severe cases after the adjustment.There were little overlapped adverse reactions during the ACT therapeutic period even in the presence of active COVID-19 infection. No death case was occurred (0/35) when those exposed to ACT regimen. Cancer patients receiving ACT had a shorter mean time to alleviation of symptoms compared with non-ACT and non-cancer (4.46 versus 16.88 and 17.90 days respectively) as well as the lowered severity incidence of symptoms (P = 0.0010). The infused ACT has not significant impact on peripheral blood count whereas the amount of CD3-CD16+CD56+ NK cells increased (P = 0.0017). The quantity of infused ACT was favorable for augmentation of possibility of severe to mild symptom shift. CONCLUSIONS These data demonstrate the clinical safety profiles while ACT infusions with active COVID-19 infection.The intervention of ACT for cancer patients could generate the benefit for symptom alleviation with improved recovery time. The concurrent ACT for advanced cancer patients during such infectious pandemic might simultaneously leverage and reduce the risk of immune compromised situation for subsequent chemotherapy complications.
Collapse
Affiliation(s)
- Congcong Li
- Department of Medical Oncology, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong New Area, Shanghai, 201399, China
| | - Dazhao Xu
- Department of Medical Oncology, Beijing Zhongguancun Hospital, Chinese Academy of Sciences, Beijing, China
| | - Linyao Lu
- Department of Medical Oncology, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong New Area, Shanghai, 201399, China
| | - Shu Peng
- Department of Medical Oncology, Fudan University Huadong Medical Center, Shanghai, China
| | - Haiyang Zhao
- Department of Medical Oncology, Beijing Zhongguancun Hospital, Chinese Academy of Sciences, Beijing, China
| | - Chuxiong Zeng
- Department of Medical Oncology, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong New Area, Shanghai, 201399, China
| | - Lina Hu
- Department of Medical Oncology, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong New Area, Shanghai, 201399, China
| | - Xianzhi Guo
- Department of Medical Oncology, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong New Area, Shanghai, 201399, China
| | - Li Liu
- Department of Medical Oncology, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong New Area, Shanghai, 201399, China
| | - Feifei Huo
- Department of Medical Oncology, Beijing Zhongguancun Hospital, Chinese Academy of Sciences, Beijing, China
| | - Xiumei Rong
- Department of Medical Oncology, Beijing Zhongguancun Hospital, Chinese Academy of Sciences, Beijing, China
| | - Zhenying Geng
- Department of Medical Oncology, Beijing Zhongguancun Hospital, Chinese Academy of Sciences, Beijing, China
| | - Ping Lin
- Department of Medical Oncology, Beijing Zhongguancun Hospital, Chinese Academy of Sciences, Beijing, China
| | - Xinna Zhou
- Center of Clinical Trials & Management, Beijing Shijitan Hospital, Capital Medical University Cancer Center, Beijing, 10038, China
| | - Xiaoli Wang
- Department of Medical Oncology, Beijing Shijitan Hospital, Capital Medical University Cancer Center, Beijing, 10038, China
| | - Amy Hobeika
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Michael A Morse
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | | | - Jun Ren
- Department of Medical Oncology, Fudan University Pudong Medical Center, 2800 Gongwei Road, Pudong New Area, Shanghai, 201399, China.
- Department of Medical Oncology, Beijing Zhongguancun Hospital, Chinese Academy of Sciences, Beijing, China.
- Department of Medical Oncology, Fudan University Huadong Medical Center, Shanghai, China.
- Department of Surgery, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
24
|
Wang J, Gong P, Liu Q, Wang M, Wu D, Li M, Zheng S, Wang H, Long Q. Stimulation of regulatory dendritic cells suppresses cytotoxic T cell function and alleviates DEN-induced liver injury, fibrosis and hepatocellular carcinoma. Front Immunol 2025; 16:1565486. [PMID: 40264769 PMCID: PMC12011597 DOI: 10.3389/fimmu.2025.1565486] [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: 01/23/2025] [Accepted: 03/06/2025] [Indexed: 04/24/2025] Open
Abstract
Background Dendritic cells (DCs) are versatile professional antigen-presenting cells and play an instrumental role in the generation of antigen-specific T-cell responses. Modulation of DC function holds promise as an effective strategy to improve anti-tumor immunotherapy efficacy and enhance self-antigen tolerance in autoimmune diseases. Methods Wild-type (WT) and TLR2 knockout (KO) mice at 2 weeks of age were injected intraperitoneally (i.p.) with a single dose of diethylnitrosamine (DEN) to induce hepatocellular carcinoma (HCC). Four weeks later, WT and KO mice were randomly divided into control and treatment groups and treated once every two days for 30 weeks with phosphate buffered saline (PBS) and a mix of 4 TLR2-activating lactic acid-producing probiotics (LAP), respectively. Mice were euthanized after 30 weeks of LAP treatment and their liver tissues were collected for gene expression, histological, flow cytometric and single-cell RNA sequencing analyses. Results We demonstrate here that oral administration of a mix of TLR2-activating LAP triggers a marked accumulation of regulatory DCs (rDCs) in the liver of mice. LAP-treated mice are protected from DEN-induced liver injury, fibrosis and HCC in a TLR2-dependent manner. Single-cell transcriptome profiling revealed that LAP treatment determines an immunosuppressive hepatic T-cell program that is characterized by a significantly reduced cytotoxic activity. The observed functional changes of T cells correlated well with the presence of a hepatic DC subset displaying a regulatory or tolerogenic transcriptional signature. Conclusion Overall, these data suggest that stimulation of regulatory dendritic cells (rDCs) in the liver by LAP suppresses cytotoxic T-cell function and alleviates DEN-induced liver damage, fibrosis and tumorigenesis.
Collapse
Affiliation(s)
- Junjie Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cam-Su Mouse Genomic Resources Center, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Pixu Gong
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cam-Su Mouse Genomic Resources Center, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Qingqing Liu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cam-Su Mouse Genomic Resources Center, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Menglei Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cam-Su Mouse Genomic Resources Center, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Dengfang Wu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cam-Su Mouse Genomic Resources Center, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Mengyu Li
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cam-Su Mouse Genomic Resources Center, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Shujie Zheng
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cam-Su Mouse Genomic Resources Center, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Han Wang
- Center for Circadian Clocks, Soochow University, Suzhou, Jiangsu, China
- School of Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Qiaoming Long
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cam-Su Mouse Genomic Resources Center, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
25
|
Liang X, Luo B, Lin B, Liu D, Guo J, Lu W, Tian S, Cai Z, Zhou X, Jin Z, Li T, Chen K, Zhou H, Wang L. Characteristics of second primary malignancies following bispecific antibodies therapy. J Immunother Cancer 2025; 13:e011200. [PMID: 40187754 PMCID: PMC11973794 DOI: 10.1136/jitc-2024-011200] [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: 12/02/2024] [Accepted: 03/24/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND The risk of secondary primary malignancies (SPMs) associated with bispecific antibody (BsAb)-a promising alternative to chimeric antigen receptor (CAR)-T therapy-remains insufficiently explored. METHODS Using large-scale, real-world data from the US Food and Drug Administration's Adverse Event Reporting System, we identified the relative frequency and characteristics of SPMs following BsAbs therapy and conducted a comprehensive comparison of treatment-related SPM profiles between BsAbs and CAR-T therapies. RESULTS We identified 108 cases among 10,280 BsAb-treated patients. The incidence risk of SPMs was stable over the past 8 years, accounting for 1-2% of all adverse events, with a case fatality rate of 29.63% among the SPM cases. Myeloid leukemias and non-Hodgkin's lymphoma were more frequent in blinatumomab recipients, while solid malignancies predominated in those treated with teclistamab. Time-to-onset (TTO) was significantly shorter in BsAb recipients compared with non-recipients, with weight and treatment duration influencing TTO, while no significant differences in TTO were observed across different BsAb products, ages, and genders. Our findings highlight the first year of BsAbs as a critical window for early detection and intervention. Although the overall risk of SPMs was lower with BsAbs than with CAR-T, the outcomes of SPMs were comparable in both groups. TTO and SPM patterns were statistically similar between the two therapies. CONCLUSION Our study provides the first detailed characterization of SPMs post-BsAb, underscoring the need for continued pharmacovigilance and individualized risk management to mitigate SPM risks in patients undergoing BsAb therapy.
Collapse
Affiliation(s)
- Xiaojie Liang
- Department of Hematology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Baiwei Luo
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Bingyu Lin
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Dan Liu
- Department of Radiology, Shunde Hospital, Southern Medical University, Foshan, China
| | - Jia Guo
- Department of Hematology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Weixiang Lu
- Department of Hematology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Shengyu Tian
- Department of Hematology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Zihong Cai
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xinyu Zhou
- The First School of Clinical Medicine, Guangdong Medical University, Zhanjiang, China
| | - Zhihao Jin
- The First School of Clinical Medicine, Guangdong Medical University, Zhanjiang, China
| | - Tong Li
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, China
| | - Keren Chen
- Department of General Medicine, Maoming People's Hospital, Maoming, China
| | - Hongsheng Zhou
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Liang Wang
- Department of Hematology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
26
|
Ton Nu QC, Deka G, Park PH. CD8 + T cell-based immunotherapy: Promising frontier in human diseases. Biochem Pharmacol 2025; 237:116909. [PMID: 40179991 DOI: 10.1016/j.bcp.2025.116909] [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: 12/10/2024] [Revised: 02/28/2025] [Accepted: 03/26/2025] [Indexed: 04/05/2025]
Abstract
The abundant cell components of the adaptive immune system called T lymphocytes (T cells) play important roles in mediating immune responses to eliminate the invaders and create the memory of the germs to form a new immunity for the next encounter. Among them, cytotoxic T cells expressing cell-surface CD8 are the most critical effector cells that directly eradicate the target infected cells by recognizing antigens presented by major histocompatibility complex class I molecules to protect our body from pathological threats. In the continuous evolution of immunotherapy, various CD8+ T cell-based therapeutic strategies have been developed based on the role and molecular concept of CD8+ T cells. The emergence of such remarkable therapies provides promising hope for multiple human disease treatments such as autoimmunity, infectious disease, cancer, and other non-infectious diseases. In this review, we aim to discuss the current knowledge on the utilization of CD8+ T cell-based immunotherapy for the treatment of various diseases, the molecular basis involved, and its limitations. Additionally, we summarize the recent advances in the use of CD8+ T cell-based immunotherapy and provide a comprehensive overview of CD8+ T cells, including their structure, underlying mechanism of function, and markers associated with CD8+ T cell exhaustion. Building upon these foundations, we delineate the advancement of CD8+ T cell-based immunotherapies with fundamental operating principles followed by research studies, and challenges, as well as illustrate human diseases involved in this development.
Collapse
Affiliation(s)
- Quynh Chau Ton Nu
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea
| | - Gitima Deka
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea
| | - Pil-Hoon Park
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea; Research institute of cell culture, Yeungnam University, Gyeongsan, Republic of Korea.
| |
Collapse
|
27
|
Shentu J, Xu H, Zhu F. Ammonia-induced lysosomal and mitochondrial damage: a novel perspective on T cell-based cancer immunotherapy. JOURNAL OF THE NATIONAL CANCER CENTER 2025; 5:105-107. [PMID: 40265095 PMCID: PMC12010380 DOI: 10.1016/j.jncc.2025.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 04/24/2025] Open
Affiliation(s)
- Jianqiao Shentu
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China
- Department of Clinical Medicine, Hangzhou City University, Hangzhou, China
| | - Hening Xu
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China
- Department of Clinical Medicine, Hangzhou City University, Hangzhou, China
| | - Feng Zhu
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, China
- Department of Clinical Medicine, Hangzhou City University, Hangzhou, China
| |
Collapse
|
28
|
Cao C, Lai H, Shi Y. MYCN/MNX1 Axis Drives NSCLC Progression by Inducing Macrophage M2 Polarization and CD8 + T Cell Apoptosis via the Wnt/β-Catenin Pathway. J Biochem Mol Toxicol 2025; 39:e70251. [PMID: 40226978 DOI: 10.1002/jbt.70251] [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: 12/03/2024] [Revised: 02/28/2025] [Accepted: 03/31/2025] [Indexed: 04/15/2025]
Abstract
Enhanced macrophage M2 polarization and CD8+ T cell dysfunction contribute to the pathophysiology of non-small cell lung cancer (NSCLC). Motor neuron and pancreatic homeobox 1 (MNX1) has emerged as a potential tumor-promoting player. Here, we clarified the activity of MNX1 in NSCLC. PMA-induced THP-1 M0-like macrophages or CD8+ T cells were co-cultured with NSCLC cells. Cell colony formation, migration, proliferation, apoptosis, and invasiveness were assessed by colony formation, wound healing, CCK-8, flow cytometry, and transwell assays, respectively. The ratio of CD206+ macrophages was analyzed by flow cytometry. Ki-67 expression was tested by immunofluorescence. ChIP and luciferase assays were used to evaluate the relationship between MYCN and MNX1. MNX1 was highly expressed in NSCLC, and its loss-of-function suppressed cell growth, motility, and invasiveness in NSCLC cells. MNX1 depletion also diminished macrophage M2 polarization and CD8+ T cell apoptosis. Mechanistically, MYCN increased MNX1 expression at the transcriptional level. MNX1 increase reversed the impact of MYCN depletion on NSCLC cell malignant behaviors, macrophage M2 polarization, and CD8+ T cell viability. MYCN depletion diminished the in vivo growth of A549 subcutaneous xenografts. Additionally, MNX1 increase counteracted the impact of MYCN depletion on the Wnt/β-catenin pathway. Our findings elucidate the oncogenic role of the MYCN/MNX1/Wnt/β-catenin pathway in NSCLC by driving macrophage M2 polarization and diminishing CD8+ T cell viability. Our study thus uncovers a novel mechanism underlying NSCLC development and highlights potential targets for combating NSCLC.
Collapse
Affiliation(s)
- Chengzhang Cao
- Department of Thoracic Surgery, Longyan First Affiliated Hospital of Fujian Medical Univensity, Longyan, China
| | - Haiyin Lai
- Department of Thoracic Surgery, Longyan First Affiliated Hospital of Fujian Medical Univensity, Longyan, China
| | - Yuzhen Shi
- Department of Critical Care Medicine, Longyan First Affiliated Hospital of Fujian Medical Univensity, Longyan, China
| |
Collapse
|
29
|
Perdomo-Celis F, Passaes C, Monceaux V, Lambotte O, Costagliola D, Chevalier MF, Weiss L, Sáez-Cirión A. Impact of rosuvastatin on the memory potential and functionality of CD8 + T cells from people with HIV. EBioMedicine 2025; 114:105672. [PMID: 40158388 PMCID: PMC11995788 DOI: 10.1016/j.ebiom.2025.105672] [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: 12/02/2024] [Revised: 03/12/2025] [Accepted: 03/14/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND Virus-specific CD8+ T cells play a major role in the natural control of HIV infection, linked to memory-like features such as high survival capacity and polyfunctionality. However, virus-specific CD8+ T cells from HIV non-controllers exhibit an effector-like and exhausted profile, with limited antiviral potential. Metabolic reprogramming of cells from non-controllers could reinvigorate their functional capacities. Considering the implication of the cholesterol pathway in the induction of T cell exhaustion, here we evaluated the impact of rosuvastatin, an inhibitor of cholesterol synthesis, on the functionality and memory profile of HIV-specific CD8+ T cells from people on antiretroviral treatment. METHODS We analysed samples from 10 individuals with HIV-1 on ART who participated in the IMEA 043-CESAR trial and received rosuvastatin for 12 weeks. We explored whether rosuvastatin treatment was accompanied by changes in the memory potential of CD8+ T cells. We evaluated the phenotype and functionality of total and HIV-specific CD8+ T cells before, during, and after treatment with rosuvastatin. A mixed effects model was used for repeated measures and corrected for multiple comparisons. FINDINGS Total and HIV-specific CD8+ T cell survival and functionality were enhanced in individuals who received a 12-week course of rosuvastatin, with a consistent increase in polyfunctional IFN-γ+ TNF-α+ cells. The superior CD8+ T cell functionality after rosuvastatin treatment was associated with intrinsic metabolic changes, including the decrease of fatty acid uptake, as well as a reduction in effector/exhaustion markers. Changes in the characteristics of CD8+ T cells coincided with the duration of rosuvastatin administration, and most effects waned after the cessation of the treatment. INTERPRETATION CD8+ T cell metabolic reprogramming by targeting the cholesterol pathway, combined with other available immunotherapies, might represent a promising strategy in the search for the cure of HIV or other chronic viral infections. FUNDING The CESAR trial was sponsored by IMEA. This work was supported by the NIH (grants UM1AI164562 and R01DK131476).
Collapse
Affiliation(s)
- Federico Perdomo-Celis
- Institut Pasteur, Université Paris Cité, Viral Reservoirs and Immune Control Unit, Paris, 75015, France; Institut Pasteur, Université Paris Cité, HIV Inflammation and Persistance Unit, Paris, 75015, France; Instituto de Genética Humana, Facultad de Medicina, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Caroline Passaes
- Institut Pasteur, Université Paris Cité, Viral Reservoirs and Immune Control Unit, Paris, 75015, France; Institut Pasteur, Université Paris Cité, HIV Inflammation and Persistance Unit, Paris, 75015, France
| | - Valérie Monceaux
- Institut Pasteur, Université Paris Cité, Viral Reservoirs and Immune Control Unit, Paris, 75015, France; Institut Pasteur, Université Paris Cité, HIV Inflammation and Persistance Unit, Paris, 75015, France
| | - Olivier Lambotte
- Université Paris-Saclay, Inserm, CEA, Center for Immunology of Viral, Auto-immune, Hematological, Bacterial Diseases (IMVA-HB/IDMIT/UMRS1184), Le Kremlin Bicêtre, Fontenay aux Roses, France; Assistance Publique Hôpitaux de Paris, Groupe Hospitalier Universitaire Paris Saclay, Service de Médecine interne immunologie clinique, Le Kremlin Bicêtre, France
| | - Dominique Costagliola
- Sorbonne Université, INSERM, Institut Pierre Louis d'Épidémiologie et de Santé Publique (IPLESP), Paris, France
| | - Mathieu F Chevalier
- INSERM UMR 1342, Institut de Recherche Saint-Louis (IRSL), Université Paris Cité, Paris, France
| | - Laurence Weiss
- Université de Paris Cité, AP-HP, Paris Centre, Paris, France
| | - Asier Sáez-Cirión
- Institut Pasteur, Université Paris Cité, Viral Reservoirs and Immune Control Unit, Paris, 75015, France; Institut Pasteur, Université Paris Cité, HIV Inflammation and Persistance Unit, Paris, 75015, France.
| |
Collapse
|
30
|
Deng J, Zhu J, Jiang X, Yao C, Chen H, Ding Y, Niu P, Chen Q, Ding H, Shen N. PD-1 activation mitigates lupus nephritis by suppressing hyperactive and heterogeneous PD-1 +CD8 + T cells. Theranostics 2025; 15:5029-5044. [PMID: 40303350 PMCID: PMC12036892 DOI: 10.7150/thno.107418] [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: 11/22/2024] [Accepted: 03/23/2025] [Indexed: 05/02/2025] Open
Abstract
Rationale: Programmed cell death protein 1 (PD-1)-expressing CD8+ T cells are typically associated with exhaustion in cancer and infections, but their role in autoimmune diseases, particularly lupus nephritis (LN), remains less understood. Understanding the characteristics and functions of PD-1+CD8+ T cells in LN could help identify novel therapeutic strategies. Methods: We analyzed the abundance and phenotypes of PD-1+CD8+ T cells in LN patients and NZB/W F1 mice. Single-cell RNA sequencing (scRNA-seq) was used to delineate subsets and TCR clonal diversity in PD-1+CD8+ T cells in NZB/W F1 mice. The therapeutic efficacy of a PD-L1 Fc fusion protein on kidney pathology and proteinuria in NZB/W F1 mice was evaluated. In addition, the inhibitory mechanism of PD-1 in CD8+ T cells were further explored using RNA-seq, q-PCR, flow cytometry, and Western blot. Results: PD-1+CD8+ T cells were enriched in LN patients and NZB/W F1 mice, exhibiting elevated activation markers and cytotoxic molecules compared to PD-1- cells. scRNA-seq identified seven distinct subsets with diverse effector functions and robust TCR clonal expansion in the kidney of NZB/W F1 mice with severe disease. PD-L1 Fc treatment reduced kidney damage and proteinuria in NZB/W F1 mice, which correlated with decreased frequencies of PD-1+CD8+ and IFN-γ+CD8+ T cells. Mechanistically, PD-L1 Fc inhibited Stat1 phosphorylation, T-bet expression, and IFN-γ production in CD8+ T cells. Conclusion: These findings show that PD-1+CD8+ T cells in LN are hyperactive, clonally expanded, and contribute to disease progression. Targeting the PD-1/PD-L1 pathway with PD-L1 Fc effectively reduced kidney pathology in a murine model of LN, underscoring the potential of modulating PD-1 signaling as a treatment strategy for LN.
Collapse
Affiliation(s)
- Jun Deng
- Shanghai Institute of Rheumatology, Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junling Zhu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoyue Jiang
- Shanghai Institute of Rheumatology, Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chao Yao
- Shanghai Institute of Rheumatology, Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haifeng Chen
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanjie Ding
- Department of Rheumatology and Immunology, Huaihe Hospital of Henan University, Kaifeng, 475000, Henan, China
| | - Peng Niu
- Shanghai Institute of Rheumatology, Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qian Chen
- Department of Ophthalmology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huihua Ding
- Shanghai Institute of Rheumatology, Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Nan Shen
- Shanghai Institute of Rheumatology, Department of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Center for Autoimmune Genomics and Etiology (CAGE), Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| |
Collapse
|
31
|
Tao X, Wang Y, Xiang B, Hu D, Xiong W, Liao W, Zhang S, Liu C, Wang X, Zhao Y. Sex bias in tumor immunity: insights from immune cells. Theranostics 2025; 15:5045-5072. [PMID: 40303343 PMCID: PMC12036885 DOI: 10.7150/thno.106465] [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: 11/06/2024] [Accepted: 03/08/2025] [Indexed: 05/02/2025] Open
Abstract
Significant sex disparities have been observed in cancer incidence, treatment response to immunotherapy, and susceptibility to adverse effects, affecting both reproductive and non-reproductive organ cancers. While lifestyle factors, carcinogenic exposure, and healthcare access contribute to these disparities, they do not fully explain the observed male-female variation in anti-tumor immunity. Despite the preferential expression of sex hormone receptors in immune cells, X chromosome also contains numerous genes involved in immune function, and its incomplete inactivation may enhance anti-tumor immune responses in females. In contrast, loss or downregulation of Y-linked genes in males has been associated with an increased cancer risk. Additionally, estrogen, progesterone and androgen signaling pathways influence both innate and adaptive immune responses, contributing to sex-specific outcomes in cancer progression and therapy. Sex-biased differences are also evident in the epigenetic regulation of gene expression, cellular senescence, microbiota composition, metabolism, and DNA damage response, all of which impact anti-tumor immunity and immunotherapy treatment efficacy. In general, the combination of sex chromosomes, sex hormones, and hormone receptors orchestrates the phenotype and function of various immune cells involved in tumor immunity. However, sex disparity in each specific immune cell are context and environment dependent, considering the preferential expression of hormone receptor in immune cell and sex hormone levels fluctuate significantly across different life stages. This review aims to outline the molecular, cellular, and epigenetic changes in T cells, B cells, NK cells, DCs, neutrophils, and macrophages driven by sex chromosomes and sex hormone signaling. These insights may inform the design of sex-specific targeted therapies and leading to more individualized cancer treatment strategies.
Collapse
Affiliation(s)
- Xuerui Tao
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yiling Wang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Binghua Xiang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Dongmei Hu
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Wei Xiong
- Department of Urology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Wenjun Liao
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Shichuan Zhang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Chi Liu
- Department of Nephrology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiaoxiao Wang
- Department of Organ Transplantation, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yue Zhao
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
32
|
Davila M, Lee SB, Kang YP, Boucher J, Mandula J, Roselli E, Chang D, Jimenez R, Kotani H, Reid K, Vazquez-Martinez J, Beatty N, Goala P, Sierra-Mondragon R, Liu M, Koomen J, Nguyen J, Hussaini M, Shaw T, Wang X, Faramand R, Jain M, Locke F, Rodriguez P, Sailer C, McSain S, Hamid S, Tariq M, Wang J, Abraham-Miranda J. CAR T cell-driven induction of iNOS in tumor-associated macrophages promotes CAR T cell resistance in B cell lymphoma. RESEARCH SQUARE 2025:rs.3.rs-3481746. [PMID: 40235478 PMCID: PMC11998770 DOI: 10.21203/rs.3.rs-3481746/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Chimeric antigen receptor (CAR) T cell therapies have revolutionized B cell malignancy treatment, but subsets of patients with large B cell lymphoma (LBCL) experience primary resistance or relapse after CAR T cell treatment. To uncover tumor microenvironment (TME)-induced resistance mechanisms, we examined patients' intratumoral immune infiltrates and observed that elevated levels of immunoregulatory macrophages in pre-infusion tumor biopsies are correlated with poor clinical responses. CAR T cell-produced interferon-gamma (IFN-γ) promotes the expression of inducible nitric oxide synthase (iNOS, NOS2) in immunoregulatory macrophages, impairing CAR T cell function. Mechanistically, iNOS-expressing macrophages upregulated the p53 pathway, mediating apoptosis and cell cycle arrest in CAR T cells, while downregulating the MYC pathway involved in ribosome biogenesis and protein synthesis. Furthermore, CAR T cell metabolism is compromised by depletion of glycolytic intermediates and rewiring of the TCA cycle. Pharmacological inhibition of iNOS enhances the CAR T cell treatment efficacy in B cell tumor-bearing mice. Notably, elevated levels of iNOS+CD14+ monocytes were observed in leukaphereses of patients with non-durable response to CAR T cell therapy. These findings suggest that mitigating iNOS in tumor-associated macrophages (TAMs) by blocking IFN-γ secretion from CAR T cells will improve outcomes for LBCL patients.
Collapse
|
33
|
Liu X, Zhang X, Liu H, Fu H, Liu Y, Ge Y, Deng S, Tang Z, Mei L, Wang J, Liu X, Yang Y, Wu Z, Ji Y. Garlic-Derived Exosome-Like Nanoparticles Enhance Gut Homeostasis in Stressed Piglets: Involvement of Lactobacillus reuteri Modulation and Indole-3-propionic Acid Induction. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:7228-7243. [PMID: 40082245 DOI: 10.1021/acs.jafc.4c11506] [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: 03/16/2025]
Abstract
The occurrence of pediatric diarrhea is frequently associated with inflammatory responses, compromised barrier function, and dysbiosis in the gut. These conditions are commonly triggered by stressors, similar to postweaning diarrhea observed in piglets. Garlic-derived exosome-like nanoparticles (GELNs) hold the potential for ameliorating stress-induced diarrhea, yet supporting evidence remains scarce. Following the successful isolation of GELNs, this study employed weaned piglets as a model to evaluate the regulatory effects of GELNs on intestinal barrier integrity, mucosal inflammation, and the gut microbiota and its metabolites. Weaned Bama miniature piglets were orally administered phosphate buffer saline (PBS) or GELNs, and 1 week later, samples were collected following slaughter. Histological and molecular biological techniques were performed to examine intestinal structure, tight junction protein expression, mucin secretion, T lymphocyte infiltration, and the levels of pro-inflammatory cytokines. The composition of the gut microbiota was analyzed using 16S rRNA sequencing, while its derived metabolites were profiled via untargeted metabolomics. Subsequently, correlation analyses were performed to evaluate the associations between the microbiota and its derived metabolites, as well as between the microbiota and the key indicators of intestinal barrier function and cytokine levels in response to GELNs. The isolated GELNs exhibit typical exosome characteristics in size and morphology, alongside a rich content of proteins and RNAs. The incidence of diarrhea in weaned piglets was reduced with supplementation of GELNs at a dosage of 50 mg/kg body weight, compared to the control group. In addition, piglets receiving GELNs displayed an increase in mucin content within the tissues of the jejunum, ileum, and colon, a decrease in CD8+ T lymphocyte counts in the colon, and suppression of pro-inflammatory cytokines (IL-8 and TNF-α) levels in the mucosal layers of both the jejunum and ileum. Furthermore, 16S rRNA sequencing unveiled that GELNs reshaped the colonic microbiota in weaned piglets by augmenting beneficial bacteria, notably Lactobacillus and Lactobacillus reuteri, correlating strongly with diminished TNF-α protein levels and heightened mucin expression. Metabolite analysis demonstrated a significant increase in indole-3-propionic acid, derived from the gut microbiota, in piglets supplemented with GELNs. This increase was positively correlated with the abundance of Lactobacillus and Lactobacillus reuteri and negatively linked with the protein levels of IL-8 and TNF-α in the gut. In summary, our study demonstrates that GELNs mitigate stress-related intestinal mucosal inflammation and enhance mucin production in the gut of weaned piglets, which is potentially achieved through the optimization of gut microbiota composition, specifically by increasing the abundance of Lactobacillus and Lactobacillus reuteri, as well as via the induction of the anti-inflammatory microbial metabolite indole-3-propionic acid. The findings presented here provide essential groundwork for the future development of GELNs as a therapeutic strategy aimed at enhancing gut homeostasis disruption caused by stress in both weaned piglets and children.
Collapse
Affiliation(s)
- Xiyuan Liu
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China
| | - Xinyu Zhang
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China
| | - Haozhen Liu
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China
| | - Huiyang Fu
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China
| | - Yanan Liu
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China
| | - Yao Ge
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China
| | - Siwei Deng
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China
| | - Zhining Tang
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China
| | - Lihua Mei
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China
| | - Jiaxin Wang
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China
| | - Xuelian Liu
- State Key Laboratory of Direct-Fed Microbial Engineering, Beijing 100192, China
| | - Ying Yang
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China
| | - Zhenlong Wu
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing 100193, China
| | - Yun Ji
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China
| |
Collapse
|
34
|
Liu C, Zhang H, Zhai YY, Dong J, Zhou Y, Li H, Zhang M, Yang CL, Zhang P, Li XL, Duan RS, Du T. Phenotypic and functional dysregulations of CD8 + T Cells in myasthenia gravis. Clin Exp Med 2025; 25:96. [PMID: 40131529 PMCID: PMC11937161 DOI: 10.1007/s10238-025-01603-4] [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: 01/19/2025] [Accepted: 02/14/2025] [Indexed: 03/27/2025]
Abstract
Myasthenia Gravis (MG) is a heterogeneous autoimmune disorder characterized by fluctuating muscle weakness caused by autoantibodies targeting neuromuscular junction components. While the role of CD4 + T cells in MG is well established, the contribution of CD8 + T cells remains poorly understood. In this study, we analyze CD8 + T cells in 36 MG patients and 38 age- and gender-matched controls using flow cytometry to evaluate subset distribution, granzyme expression, and cytokine production. MG patients exhibit an altered CD4 + /CD8 + T cell ratio and significant changes in CD8 + T cell subsets, including increased central memory CD8 + T cell (Tcm) proportions and decreased effector memory CD8 + T cell (Tem) proportions. Granzyme B expression in Tcm cells is significantly elevated in MG patients, whereas no significant changes are observed in other subsets or GZMK expression. Cytokine analysis reveals increased IL-21, GM-CSF, and IL-17A production by CD8 + T cells in MG patients. These phenotypic and functional alterations of CD8 + T cells persist during the acute phase of the disease, regardless of immunotherapy usage, and vary between ocular and generalized MG. Subgroup and correlation analyses further identify age-dependent and age-independent dysregulations of CD8 + T cells, indicating complex and subtype-specific roles of CD8 + T cells in the immunopathological processes underlying MG. Our findings provide novel insights into the involvement of CD8 + T cells in MG pathogenesis, laying a foundation for future research and potential therapeutic strategies targeting CD8 + T cells.
Collapse
Affiliation(s)
- Chang Liu
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, People's Republic of China
| | - Hao Zhang
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, People's Republic of China
| | - Yu-Yao Zhai
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, People's Republic of China
| | - Jing Dong
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, People's Republic of China
| | - Yang Zhou
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, People's Republic of China
- Shandong Institute of Neuroimmunology, Jinan, 250014, People's Republic of China
- Shandong Provincial Medicine and Health Key Laboratory of Neuroimmunology, Jinan, 250014, People's Republic of China
| | - Heng Li
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, People's Republic of China
- Shandong Institute of Neuroimmunology, Jinan, 250014, People's Republic of China
- Shandong Provincial Medicine and Health Key Laboratory of Neuroimmunology, Jinan, 250014, People's Republic of China
| | - Min Zhang
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, People's Republic of China
- Shandong Institute of Neuroimmunology, Jinan, 250014, People's Republic of China
- Shandong Provincial Medicine and Health Key Laboratory of Neuroimmunology, Jinan, 250014, People's Republic of China
| | - Chun-Lin Yang
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, People's Republic of China
- Shandong Institute of Neuroimmunology, Jinan, 250014, People's Republic of China
- Shandong Provincial Medicine and Health Key Laboratory of Neuroimmunology, Jinan, 250014, People's Republic of China
| | - Peng Zhang
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, People's Republic of China
- Shandong Institute of Neuroimmunology, Jinan, 250014, People's Republic of China
- Shandong Provincial Medicine and Health Key Laboratory of Neuroimmunology, Jinan, 250014, People's Republic of China
| | - Xiao-Li Li
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, People's Republic of China
- Shandong Institute of Neuroimmunology, Jinan, 250014, People's Republic of China
- Shandong Provincial Medicine and Health Key Laboratory of Neuroimmunology, Jinan, 250014, People's Republic of China
| | - Rui-Sheng Duan
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, People's Republic of China.
- Shandong Institute of Neuroimmunology, Jinan, 250014, People's Republic of China.
- Shandong Provincial Medicine and Health Key Laboratory of Neuroimmunology, Jinan, 250014, People's Republic of China.
| | - Tong Du
- Department of Neurology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, People's Republic of China.
- Shandong Institute of Neuroimmunology, Jinan, 250014, People's Republic of China.
- Shandong Provincial Medicine and Health Key Laboratory of Neuroimmunology, Jinan, 250014, People's Republic of China.
| |
Collapse
|
35
|
Tao R, Ruan J, Chen X, Pang B, Li S, Zhou S, Aghayants S, Shi Z, Zhu Z. Development and validation of an immune signature-based risk model for prognostic assessment in melanoma. Sci Rep 2025; 15:9117. [PMID: 40097490 PMCID: PMC11914537 DOI: 10.1038/s41598-025-90917-0] [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: 09/10/2024] [Accepted: 02/17/2025] [Indexed: 03/19/2025] Open
Abstract
Melanoma is a highly invasive malignancy with poor prognoses in advanced stages. Developing a risk model that can accurately assess prognosis and guide personalized treatment is crucial for improving the clinical management of melanoma. This study aims to develop and validate an immune-based prognostic risk model for melanoma through comprehensive bioinformatics analysis. We collected transcriptomic data from multiple public databases and identified 9 immune features significantly associated with prognosis using single-sample Gene Set Enrichment Analysis (ssGSEA) and Cox regression. These features were utilized to construct the risk model, which was subsequently validated using relevant bulk transcriptomic datasets and single-cell transcriptomic datasets from the GEO database, encompassing diverse patient populations and sample types. The model effectively stratified patients into high-risk and low-risk groups with distinct survival outcomes. Further analysis revealed significant associations between the risk model and genomic heterogeneity indicators, such as tumor mutational burden (TMB), loss of heterozygosity (LOH), and immune checkpoint gene expression. The model robustness was confirmed using single-cell transcriptomic data, highlighting key genes with potential therapeutic relevance. Our findings provide a reliable prognostic tool and novel insights for personalized melanoma treatment, emphasizing the need for further clinical validation.
Collapse
Affiliation(s)
- Rui Tao
- Department of Plastic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei , China
| | - Jingjing Ruan
- Department of Burns, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, 430061, Hubei, China
| | - Xuejie Chen
- Department of Plastic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei , China
| | - Boshi Pang
- College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Sicheng Li
- Department of Plastic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei , China
| | - Shengzhi Zhou
- Department of Plastic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei , China
| | - Sis Aghayants
- Department of Plastic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei , China
| | - Zeqi Shi
- Department of Dermatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430014, Hubei, China.
| | - Zhanyong Zhu
- Department of Plastic Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei , China.
| |
Collapse
|
36
|
Wu X, Liu C, Zhang C, Kuai L, Hu S, Jia N, Song J, Jiang W, Chen Q, Li B. The Role of Lactate and Lactylation in the Dysregulation of Immune Responses in Psoriasis. Clin Rev Allergy Immunol 2025; 68:28. [PMID: 40080284 DOI: 10.1007/s12016-025-09037-2] [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] [Accepted: 02/24/2025] [Indexed: 03/15/2025]
Abstract
Historically, lactate has been considered merely a metabolic byproduct. However, recent studies have revealed that lactate plays a much more dynamic role, acting as an immune signaling molecule that influences cellular communication, through the process of "lactate shuttling." Lactylation, a novel post-translational modification, is directly derived from lactate and represents an emerging mechanism through which lactate exerts its effects on cellular function. It has been shown to directly affect immune cells by modulating the activation of pro-inflammatory and anti-inflammatory pathways. This modification influences the expression of key immune-related genes, thereby impacting immune cell differentiation, cytokine production, and overall immune response. In this review, we focused on the role of lactate and lactylation in the dysregulation of immune responses in psoriasis and its relapse. Additionally, we discuss the potential applications of targeting lactate metabolism and lactylation modifications in the treatment of psoriasis, alongside the investigation of artificial intelligence applications in advancing lactate and lactylation-focused drug development, identifying therapeutic targets, and enabling personalized medical decision-making. The significance of this review lies in its comprehensive exploration of how lactate and lactylation contribute to immune dysregulation, offering a novel perspective for understanding the metabolic and epigenetic changes associated with psoriasis. By identifying the roles of these pathways in modulating immune responses, this review provides a foundation for the development of new therapeutic strategies that target these mechanisms.
Collapse
Affiliation(s)
- Xinxin Wu
- Central Laboratory, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Changya Liu
- Longhua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Caiyun Zhang
- Central Laboratory, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Le Kuai
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Sheng Hu
- Central Laboratory, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Ning Jia
- Central Laboratory, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Jiankun Song
- Central Laboratory, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Wencheng Jiang
- Central Laboratory, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China.
| | - Qilong Chen
- Central Laboratory, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China.
| | - Bin Li
- Central Laboratory, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China.
| |
Collapse
|
37
|
Liu H, Liu T, Wang X, Zhu X, He J, Wang H, Fan A, Zhang D. Design and development of a novel multi-epitope DNA vaccine candidate against infectious bronchitis virus: an immunoinformatic approach. Arch Microbiol 2025; 207:84. [PMID: 40067376 DOI: 10.1007/s00203-025-04283-6] [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: 12/16/2024] [Revised: 02/10/2025] [Accepted: 02/21/2025] [Indexed: 03/27/2025]
Abstract
Avian infectious bronchitis (IB) is one of the major respiratory diseases in poultry. At present, attenuated vaccines are the main commercial vaccines, but they have many defects. We aimed to construct a novel multi-epitope DNA vaccine based on avian infectious bronchitis virus (IBV) S1 and N proteins for the prevention of IBV infection. We screened the dominant B and T cell epitopes of target proteins utilizing epitope prediction tools. A new high-immunogenicity epitope peptide segment named QSN was designed and screened by linking peptide. The physicochemical properties of QSN were analyzed by bioinformatics. The recombinant plasmid pEGFP-QSN was obtained by inserting the synthesized QSN gene into the eukaryotic expression vector pEGFP-N1. On the 7th day of age, chicks were immunized by intramuscular injection of the plasmid, and serum specific antibody IgG, cytokines IFN-γ and IL-2, and T lymphocyte subsets were detected after booster immunization. Bioinformatics analysis showed that QSN had high hydrophilicity without transmembrane region and stable structure after binding to receptor. The recombinant eukaryotic vector was successfully constructed. Two weeks after booster immunization, compared with NS group and pEGFP-N1 group, serum IgG level, concentrations of cytokines IFN-γ and IL-2, and proportion of CD4+ T lymphocytes in pEGFP-QSN group were significantly increased (P < 0.01 or P < 0.05). Collectively, the multi-epitope DNA could stimulate humoral and cellular immune responses in chickens and is expected to be a potential vaccine candidate against IBV infection.
Collapse
MESH Headings
- Animals
- Vaccines, DNA/immunology
- Vaccines, DNA/genetics
- Vaccines, DNA/administration & dosage
- Infectious bronchitis virus/immunology
- Infectious bronchitis virus/genetics
- Chickens
- Viral Vaccines/immunology
- Viral Vaccines/genetics
- Viral Vaccines/administration & dosage
- Poultry Diseases/prevention & control
- Poultry Diseases/virology
- Poultry Diseases/immunology
- Coronavirus Infections/prevention & control
- Coronavirus Infections/veterinary
- Coronavirus Infections/immunology
- Coronavirus Infections/virology
- Antibodies, Viral/blood
- Epitopes, T-Lymphocyte/immunology
- Epitopes, T-Lymphocyte/genetics
- Computational Biology
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/genetics
- Epitopes, B-Lymphocyte/immunology
- Epitopes, B-Lymphocyte/genetics
Collapse
Affiliation(s)
- Haoyu Liu
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, No. 22, Jinjing Road, Xiqing District, Tianjin, 300392, People's Republic of China
| | - Tingting Liu
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, No. 22, Jinjing Road, Xiqing District, Tianjin, 300392, People's Republic of China
| | - Xinyuan Wang
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, No. 22, Jinjing Road, Xiqing District, Tianjin, 300392, People's Republic of China
| | - Xiaochen Zhu
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, No. 22, Jinjing Road, Xiqing District, Tianjin, 300392, People's Republic of China
| | - Jinling He
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, No. 22, Jinjing Road, Xiqing District, Tianjin, 300392, People's Republic of China
| | - Hui Wang
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, No. 22, Jinjing Road, Xiqing District, Tianjin, 300392, People's Republic of China
| | - Aili Fan
- Hengnuoyou (Tianjin) Biotechnology Co., Ltd, Tianjin, 301600, China
| | - Dongchao Zhang
- Tianjin Key Laboratory of Agricultural Animal Breeding and Healthy Husbandry, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, No. 22, Jinjing Road, Xiqing District, Tianjin, 300392, People's Republic of China.
| |
Collapse
|
38
|
Li K, Zhu Y, Fang Z, Geng M, Zhang J, Zheng Y, Cao Y, Wei X, Yang J. Fish requires FasL to facilitate CD8+ T-cell function and antimicrobial immunity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkaf022. [PMID: 40073091 DOI: 10.1093/jimmun/vkaf022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/22/2025] [Accepted: 01/28/2025] [Indexed: 03/14/2025]
Abstract
Although bony fish have CD8+ T cells, the mechanisms by which these early-evolved cytotoxic cells combat intracellular pathogens remain unclear. In the present study, using Nile tilapia as a model, we investigated the detailed function, mechanism, and evolutionary pattern concerning CD8+ T cells. By depleting CD8+ T cells, they are found essential in combating Edwardsiella piscicida infection. Using siRNA interference, we propose that unlike the strategy predominantly relying on perforin/granzyme in mammals, CD8+ T-cell effector function is mediated by both FasL and perforin/granzyme in fish. Upon E. piscicida infection, FasL is induced to express in CD8+ T cells; both recombinant FasL and adoptively transferred FasL+CD8+ T cells facilitate the apoptosis of target cells. Meanwhile, tilapia FasL also triggers the apoptosis of T cells to archive homeostasis. Since advances in mammals highlight the indispensable role of FasL in maintaining CD8+ T-cell homeostasis, rather than in effector function or anti-infective immunity, we therefore propose the unique dual function of FasL in executing effector function and maintaining homeostasis in fish. Mechanistically, tilapia T cells utilize mTORC1/c-Myc axis to regulate pathogen-induced FasL expression, which binds to Fas and activates caspase-8/caspase-3 pathway, mediating apoptosis in target cells and T cells themselves. This represents a novel mechanism underpinning CD8+ T-cell function in fish. Our findings demonstrate that CD8+ T cells reshaped the FasL-dependent strategy throughout evolution, thereby enhancing the precision and specificity of adaptive immunity.
Collapse
Affiliation(s)
- Kang Li
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, China
| | - Yating Zhu
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, China
| | - Zhichao Fang
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, China
| | - Ming Geng
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, China
| | - Jiansong Zhang
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, China
| | - Yuying Zheng
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, China
| | - Yi Cao
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, China
| | - Xiumei Wei
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, China
| | - Jialong Yang
- State Key Laboratory of Estuarine and Coastal Research, School of Life Sciences, East China Normal University, Shanghai, China
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China
| |
Collapse
|
39
|
Sarkar S, Taira N, Hsieh TH, Chien HC, Hirota M, Koizumi SI, Sasaki D, Tamai M, Seto Y, Miyagi M, Ishikawa H. JunB is required for CD8+ T cell responses to acute infections. Int Immunol 2025; 37:203-220. [PMID: 39425978 PMCID: PMC11884676 DOI: 10.1093/intimm/dxae063] [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: 02/26/2024] [Accepted: 10/15/2024] [Indexed: 10/21/2024] Open
Abstract
Basic-leucine zipper transcription factor ATF-like (BATF) and interferon regulatory factor 4 (IRF4) are crucial transcription factors for the generation of cytotoxic effector and memory CD8+ T cells. JunB is required for expression of genes controlled by BATF and IRF4 in CD4+ T cell responses, but the role of JunB in CD8+ T cells remains unknown. Here, we demonstrate that JunB is essential for cytotoxic CD8+ T cell responses. JunB expression is transiently induced, depending on the T cell receptor signal strength. JunB deficiency severely impairs the clonal expansion of effector CD8+ T cells in response to acute infection with Listeria monocytogenes. Junb-deficient CD8+ T cells fail to control transcription and chromatin accessibility of a specific set of genes regulated by BATF and IRF4, resulting in impaired cell survival, glycolysis, and cytotoxic CD8+ T cell differentiation. Furthermore, JunB deficiency enhances the expression of co-inhibitory receptors, including programmed cell death 1 (PD-1) and T cell immunoglobulin mucin-3 (TIM3) upon activation of naive CD8+ T cells. These results indicate that JunB, in collaboration with BATF and IRF4, promotes multiple key events in the early stage of cytotoxic CD8+ T cell responses.
Collapse
Affiliation(s)
- Shukla Sarkar
- Immune Signal Unit, Okinawa Institute of Science and Technology, Graduate University (OIST), Onna-son, Okinawa 904-0495, Japan
| | - Naoyuki Taira
- Immune Signal Unit, Okinawa Institute of Science and Technology, Graduate University (OIST), Onna-son, Okinawa 904-0495, Japan
| | - Tsung-Han Hsieh
- Immune Signal Unit, Okinawa Institute of Science and Technology, Graduate University (OIST), Onna-son, Okinawa 904-0495, Japan
| | - Hsiao-Chiao Chien
- Immune Signal Unit, Okinawa Institute of Science and Technology, Graduate University (OIST), Onna-son, Okinawa 904-0495, Japan
| | - Masato Hirota
- Immune Signal Unit, Okinawa Institute of Science and Technology, Graduate University (OIST), Onna-son, Okinawa 904-0495, Japan
| | - Shin-ichi Koizumi
- Immune Signal Unit, Okinawa Institute of Science and Technology, Graduate University (OIST), Onna-son, Okinawa 904-0495, Japan
| | - Daiki Sasaki
- Immune Signal Unit, Okinawa Institute of Science and Technology, Graduate University (OIST), Onna-son, Okinawa 904-0495, Japan
| | - Miho Tamai
- Immune Signal Unit, Okinawa Institute of Science and Technology, Graduate University (OIST), Onna-son, Okinawa 904-0495, Japan
| | - Yu Seto
- Immune Signal Unit, Okinawa Institute of Science and Technology, Graduate University (OIST), Onna-son, Okinawa 904-0495, Japan
| | - Mio Miyagi
- Immune Signal Unit, Okinawa Institute of Science and Technology, Graduate University (OIST), Onna-son, Okinawa 904-0495, Japan
| | - Hiroki Ishikawa
- Immune Signal Unit, Okinawa Institute of Science and Technology, Graduate University (OIST), Onna-son, Okinawa 904-0495, Japan
| |
Collapse
|
40
|
Qiu M, Zhao L, Li X, Fan Y, Liu M, Hua D, Zhu Y, Liang Y, Zhang Y, Xiao W, Xu X, Li J. Decoding dengue's neurological assault: insights from single-cell CNS analysis in an immunocompromised mouse model. J Neuroinflammation 2025; 22:62. [PMID: 40038739 PMCID: PMC11877810 DOI: 10.1186/s12974-025-03383-w] [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: 12/10/2024] [Accepted: 02/17/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Dengue encephalitis, a severe neurological complication of dengue virus infection, is increasingly recognized for its rising incidence and significant public health burden. Despite its growing prevalence, the underlying mechanisms and effective therapeutic strategies remain poorly understood. METHODS Cellular atlas of dengue encephalitis was determined by single-nucleus RNA sequencing. Viral load of dengue virus and the level of cytokines expression was detected by RT-qPCR. The target cells of dengue virus were verified by immunofluorescence. The cytotoxic effect of CD8+ T cell was determined by flow cytometry, immunofluorescence, in vivo CD8+ T cell depletion, adoptive transfer and CCK-8-based cell viability assay. Axonal and synaptic reduction induced by dengue virus infection was demonstrated by RT-qPCR, Western blot, transmission electron microscope and immunofluorescence. Finally, motor and sensory functions of mice were detected by open field test and hot plate test, respectively. RESULTS In this study, we utilized single-nucleus RNA sequencing on brain tissues from a dengue-infected murine model to construct a comprehensive cellular atlas of dengue encephalitis. Our findings identify neurons, particularly inhibitory GABAergic subtypes, as the primary targets of dengue virus. Additionally, immune cell infiltration was observed, contributing to significant neurological damage. Comprehensive analyses of cell-cell communication, combined with CD8+ T cell depletion and transfer restoration experiments, have elucidated the critical role of CD8+ T cells in triggering encephalitis through their interaction with neurons. These cells infiltrate the brain from peripheral circulation, interact with neurons, and induce damage of synapse and axon, accompanied by neurological dysfunction. CONCLUSION We defined cellular atlas of dengue encephalitis in mouse model and identified the primary target neuron of dengue virus. In addition, we demonstrated the significant cytotoxic effect of CD8+ T cell, which leads to apoptosis of neuron and neurological dysfunction of mice. Our study provides a molecular and cellular framework for understanding dengue encephalitis through advanced sequencing technologies. The insights gained serve as a foundation for future investigations into its pathogenesis and the development of targeted therapeutic approaches.
Collapse
Affiliation(s)
- Minyue Qiu
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
- Institute of Immunology, Army Medical University, Chongqing, China
| | - Lixin Zhao
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
- Institute of Immunology, Army Medical University, Chongqing, China
| | - Xiaojia Li
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
| | - Yipei Fan
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
| | - Minchi Liu
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
| | - Dong Hua
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
| | - Yunkai Zhu
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
| | - Yinyin Liang
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
| | - Yu Zhang
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
| | - Wen Xiao
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
| | - Xiaofeng Xu
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China
| | - Jintao Li
- Department of Biosafety, School of Basic Medicine, Army Medical University, Chongqing, China.
- Institute of Immunology, Army Medical University, Chongqing, China.
| |
Collapse
|
41
|
Pretto S, Yu Q, Bourdely P, Trusso Cafarello S, Van Acker HH, Verelst J, Richiardone E, Vanheer L, Roshanzadeh A, Schneppenheim F, Cresens C, Sassano ML, Dehairs J, Carion M, Ismail S, Agostinis P, Rocha S, Bald T, Swinnen J, Corbet C, Lunt SY, Thienpont B, Di Matteo M, Mazzone M. A functional single-cell metabolic survey identifies Elovl1 as a target to enhance CD8 + T cell fitness in solid tumours. Nat Metab 2025; 7:508-530. [PMID: 40065102 PMCID: PMC11946891 DOI: 10.1038/s42255-025-01233-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 02/04/2025] [Indexed: 03/28/2025]
Abstract
Reprogramming T cell metabolism can improve intratumoural fitness. By performing a CRISPR/Cas9 metabolic survey in CD8+ T cells, we identified 83 targets and we applied single-cell RNA sequencing to disclose transcriptome changes associated with each metabolic perturbation in the context of pancreatic cancer. This revealed elongation of very long-chain fatty acids protein 1 (Elovl1) as a metabolic target to sustain effector functions and memory phenotypes in CD8+ T cells. Accordingly, Elovl1 inactivation in adoptively transferred T cells combined with anti-PD-1 showed therapeutic efficacy in resistant pancreatic and melanoma tumours. The accumulation of saturated long-chain fatty acids in Elovl1-deficient T cells destabilized INSIG1, leading to SREBP2 activation, increased plasma membrane cholesterol and stronger T cell receptor signalling. Elovl1-deficient T cells increased mitochondrial fitness and fatty acid oxidation, thus withstanding the metabolic stress imposed by the tumour microenvironment. Finally, ELOVL1 in CD8+ T cells correlated with anti-PD-1 response in patients with melanoma. Altogether, Elovl1 targeting synergizes with anti-PD-1 to promote effective T cell responses.
Collapse
Affiliation(s)
- Samantha Pretto
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Qian Yu
- Laboratory for Functional Epigenetics, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Pierre Bourdely
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Sarah Trusso Cafarello
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Heleen H Van Acker
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Joren Verelst
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Elena Richiardone
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Brussels, Belgium
| | - Lotte Vanheer
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Amir Roshanzadeh
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
| | - Franziska Schneppenheim
- Institute of Experimental Oncology (IEO), University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Charlotte Cresens
- Molecular Imaging and Photonics Division, Chemistry Department, Faculty of Sciences, KU Leuven, Heverlee, Belgium
- VIB BioImaging Core, Leuven, Belgium
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
| | - Maria Livia Sassano
- Cell Death Research and Therapy Group, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
- VIB Center for Cancer Biology, Leuven, Belgium
| | - Jonas Dehairs
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Martin Carion
- Department of Chemistry, KU Leuven, Heverlee, Belgium
| | - Shehab Ismail
- Department of Chemistry, KU Leuven, Heverlee, Belgium
| | - Patrizia Agostinis
- Cell Death Research and Therapy Group, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
- VIB Center for Cancer Biology, Leuven, Belgium
| | - Susana Rocha
- Molecular Imaging and Photonics Division, Chemistry Department, Faculty of Sciences, KU Leuven, Heverlee, Belgium
| | - Tobias Bald
- Institute of Experimental Oncology (IEO), University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Johan Swinnen
- Laboratory of Lipid Metabolism and Cancer, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Cyril Corbet
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Brussels, Belgium
| | - Sophia Y Lunt
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
- Department of Chemical Engineering and Materials Science, Michigan State University, East Lansing, MI, USA
| | - Bernard Thienpont
- Laboratory for Functional Epigenetics, Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Mario Di Matteo
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Massimiliano Mazzone
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, VIB, Leuven, Belgium.
- Laboratory of Tumor Inflammation and Angiogenesis, Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven, Belgium.
| |
Collapse
|
42
|
Wu J, Qian P, Han Y, Xu C, Xia M, Zhan P, Wei J, Dong J. GLP1 alleviates oleic acid-propelled lipocalin-2 generation by tumor-infiltrating CD8 + T cells to reduce polymorphonuclear MDSC recruitment and enhances viral immunotherapy in pancreatic cancer. Cell Mol Immunol 2025; 22:282-299. [PMID: 39910336 PMCID: PMC11868399 DOI: 10.1038/s41423-025-01260-3] [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: 08/01/2024] [Accepted: 01/12/2025] [Indexed: 02/07/2025] Open
Abstract
Recruitment of polymorphonuclear MDSCs (PMN-MDSCs) in the TME suppresses the antitumor activity of tumor-infiltrating CD8+ T cells (CD8+ TILs). Little is known about the role of antitumoral CD8+ TILs in actively initiating an immune-tolerant microenvironment, particularly in the recruitment of PMN-MDSCs. In this study, we found that immunotherapy-activated CD8+ TILs significantly increased PNM-MDSC infiltration in the TME, resulting in antitumor resistance. When CD8+ T cells are activated, lipocalin-2 (LCN2) expression is strongly upregulated, which significantly enhances PMN-MDSC chemotaxis. Mechanistically, immune activation increased fatty acid synthesis in CD8+ T cells, particularly oleic acid (OA), which induced lysosomal membrane permeabilization, releasing cathepsin B and subsequently activating NF-κB to promote LCN2 expression. Moreover, we showed that glucagon-like peptide 1 (GLP1) effectively inhibited OA synthesis in activated CD8+ T cells, reducing LCN2 production. We then developed a recombinant adenovirus encoding GLP1 (AdV-GLP1), which significantly reduced PMN-MDSC infiltration and reinvigorated the antitumor activity of CD8+ TILs. In various pancreatic cancer models, including subcutaneous, orthotopic, and humanized CDX/PDX models, AdV-GLP1 displayed excellent antitumor efficacy. Our work advances the understanding of how immunotherapy-activated CD8+ TILs initiate PMN-MDSC infiltration and provides a clinically relevant strategy to target this interaction and improve cancer immunotherapy.
Collapse
Affiliation(s)
- Jingyi Wu
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Peng Qian
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Yifeng Han
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Chuning Xu
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Mao Xia
- Department of Clinical Laboratory Medicine, the Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Ping Zhan
- Department of Respiratory Medicine, Jinling Hospital, Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Jiwu Wei
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, China.
| | - Jie Dong
- State Key Laboratory of Pharmaceutical Biotechnology and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, Jiangsu, China.
- Department of Pathogen Biology, School of Medicine, Nantong University, Nantong, Jiangsu, 226001, China.
| |
Collapse
|
43
|
Huang W, Wang J, Liu C, Yang C, Chen Z, Ding J, Jiang W, Wang Y, Meng Y, Li L, Liu Y, Liu X, Li H, Sun B. Norepinephrine promotes activated B cells to identify and kill effector CD8 + T cells through FasL/Fas pathway in spleen mononuclear cells isolated from experimental autoimmune encephalomyelitis. Brain Behav Immun 2025; 125:294-307. [PMID: 39824471 DOI: 10.1016/j.bbi.2025.01.007] [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: 05/23/2024] [Revised: 12/18/2024] [Accepted: 01/13/2025] [Indexed: 01/20/2025] Open
Abstract
It has been reported that the nervous system can regulate immune reactions through various mechanisms. However, the role of splenic sympathetic nerve activity in the autoimmune reactions during the pathogenesis of experimental autoimmune encephalomyelitis (EAE) remained unclear. Here, we blocked the activity of the splenic sympathetic nerve and found that the number of adaptive immune cells, such as CD4+ T cells, CD8+ T cells and B cells, were upregulated. Additionally, there was an increase in the secretion of inflammatory cytokines in the spleen, and the neurological symptoms of EAE were exacerbated. In vitro experiments, we found that norepinephrine (NE), the neurotransmitter of the splenic sympathetic nerve, indirectly drove the death of effector CD8+ T cells. Furthermore, activated B cells, under the influence of NE, specifically recognized effector CD8+ T cells by upregulating MHC-I molecules and killed these cells via the FasL/Fas pathway. Our findings provide a new perspective on B cells killing effect in vitro, which was boosted by NE and demonstrate that the splenic sympathetic nerve controls the degree of autoimmune responses in EAE. This adds a new dimension to the diversity of NE's regulatory effects on adaptive immune cells and suggests a potential new therapeutic approach for autoimmune diseases.
Collapse
Affiliation(s)
- Wei Huang
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, Heilongjiang, PR China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin 150081, Heilongjiang, PR China
| | - Jing Wang
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, Heilongjiang, PR China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin 150081, Heilongjiang, PR China
| | - Chao Liu
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, Heilongjiang, PR China
| | - Changxin Yang
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, Heilongjiang, PR China
| | - Zhengyi Chen
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, Heilongjiang, PR China
| | - Jianwen Ding
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, Heilongjiang, PR China
| | - Wenkang Jiang
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, Heilongjiang, PR China
| | - Yanping Wang
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, Heilongjiang, PR China
| | - Yanting Meng
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, Heilongjiang, PR China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin 150081, Heilongjiang, PR China
| | - Lei Li
- Department of Neurology, Second Affiliated Hospital of Harbin Medical University, Harbin 150086, Heilongjiang, PR China
| | - Yumei Liu
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, Heilongjiang, PR China
| | - Xijun Liu
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, Heilongjiang, PR China
| | - Hulun Li
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, Heilongjiang, PR China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin 150081, Heilongjiang, PR China.
| | - Bo Sun
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin 150081, Heilongjiang, PR China; The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin 150081, Heilongjiang, PR China.
| |
Collapse
|
44
|
Lyu T, Wu K, Zhou Y, Kong T, Li L, Wang K, Fu P, Wei P, Chen M, Zheng J. Single-Cell RNA Sequencing Reveals the Tumor Heterogeneity and Immunosuppressive Microenvironment in Urothelial Carcinoma. Cancer Sci 2025; 116:710-723. [PMID: 39726326 PMCID: PMC11875766 DOI: 10.1111/cas.16436] [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: 07/12/2024] [Revised: 11/24/2024] [Accepted: 12/09/2024] [Indexed: 12/28/2024] Open
Abstract
Urothelial carcinoma (UC) can arise from either the lower urinary tract or the upper tract; they represent different disease entities and require different clinical treatment strategies. A full understanding of the cellular characteristics in UC may guide the development of novel therapies. Here, we performed single-cell transcriptome analysis from four patients with UC of the bladder (UCB), five patients with UC of the ureter (UCU), and four patients with UC of the renal pelvis (UCRP) to develop a comprehensive cell atlas of UC. We found the rare epithelial cell subtype EP9 with epithelial-to-mesenchymal transition (EMT) and cancer stem cell (CSC) features, and specifically expressed SOX6, which was associated with poor prognosis. We also found that ACKR1+ endothelial cells and inflammatory cancer-associated fibroblasts (iCAFs) were more enriched in UCU, which may promote pathogenesis. While ESM1+ endothelial cells may more actively participate in UCB and UCRP tumorigenesis by promoting angiogenesis. Additionally, CD8 + effector T cells were more enriched in UCU and UCRP patients, while Tregs were mainly enriched in UCB tumors. C1QC+ macrophages and LAMP3+ dendritic cells were more enriched in UCB, which is closely related to the formation of the heterogeneous immunosuppressive microenvironment. Furthermore, we found strong interactions between iCAFs, EP9, and Endo_ESM1, and different degrees of activation of the FGF-FGFR3 axis and immune checkpoint pathway were observed in different UC subtypes. Our study elucidated the cellular heterogeneity and the components of the microenvironment in UC arising from the upper and lower urinary tracts and provided novel therapeutic targets.
Collapse
Affiliation(s)
- Tianqi Lyu
- Cixi Institute of Biomedical Engineering, Chinese Academy of Science (CAS)Ningbo Institute of Materials Technology and Engineering, CAS NingboNingboChina
| | - Kerong Wu
- Department of Urology, Ningbo First HospitalSchool of Medicine Ningbo University, Zhejiang University Ningbo HospitalNingboChina
| | - Yincong Zhou
- Department of Bioinformatics, College of Life SciencesZhejiang UniversityHangzhouChina
| | - Tong Kong
- Cixi Institute of Biomedical Engineering, Chinese Academy of Science (CAS)Ningbo Institute of Materials Technology and Engineering, CAS NingboNingboChina
| | - Lin Li
- Cixi Institute of Biomedical Engineering, Chinese Academy of Science (CAS)Ningbo Institute of Materials Technology and Engineering, CAS NingboNingboChina
| | - Kaizhe Wang
- Cixi Institute of Biomedical Engineering, Chinese Academy of Science (CAS)Ningbo Institute of Materials Technology and Engineering, CAS NingboNingboChina
| | - Pan Fu
- Cixi Institute of Biomedical Engineering, Chinese Academy of Science (CAS)Ningbo Institute of Materials Technology and Engineering, CAS NingboNingboChina
| | - Pengyao Wei
- Cixi Institute of Biomedical Engineering, Chinese Academy of Science (CAS)Ningbo Institute of Materials Technology and Engineering, CAS NingboNingboChina
| | - Ming Chen
- Department of Bioinformatics, College of Life SciencesZhejiang UniversityHangzhouChina
| | - Jianping Zheng
- Cixi Institute of Biomedical Engineering, Chinese Academy of Science (CAS)Ningbo Institute of Materials Technology and Engineering, CAS NingboNingboChina
| |
Collapse
|
45
|
Poch A, Utzschneider DT. Nutrient metabolism shapes epigenetic landscape of T cells. Nat Immunol 2025; 26:340-341. [PMID: 39891020 DOI: 10.1038/s41590-025-02080-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2025]
Affiliation(s)
- Annika Poch
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Daniel T Utzschneider
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
46
|
Mori S, Fujiwara-Tani R, Ogata R, Ohmori H, Fujii K, Luo Y, Sasaki T, Nishiguchi Y, Bhawal UK, Kishi S, Kuniyasu H. Anti-Cancer and Pro-Immune Effects of Lauric Acid on Colorectal Cancer Cells. Int J Mol Sci 2025; 26:1953. [PMID: 40076581 PMCID: PMC11901037 DOI: 10.3390/ijms26051953] [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: 01/12/2025] [Revised: 02/19/2025] [Accepted: 02/22/2025] [Indexed: 03/14/2025] Open
Abstract
Lauric acid (LAA) is a 12-carbon medium-chain fatty acid that reportedly has antitumor and muscle-protecting effects. However, the details of these antitumor effects remain unclear. Therefore, in this study, we investigated the mechanism underlying the antitumor effects of LAA in CT26 and HT29 colorectal cancer (CRC) cell lines. Our in vitro findings demonstrated that LAA suppressed CRC cell proliferation, induced mitochondrial oxidative stress (reactive oxygen species (ROS)), inhibited oxidative phosphorylation (OXPHOS), and induced apoptosis. Moreover, in vivo analysis of LAA showed a more pronounced antitumor effect in CT26 cells in a syngeneic mouse tumor model than in vitro; therefore, we further investigated its impact on host antitumor immunity. We observed that LAA increased the number of effector T cells in mouse tumors, while in vitro LAA activated mouse splenocytes (SplC) and promoted OXPHOS. In two-dimensional co-culture of SplC and CT26 cells, LAA induced cell death in cancer cells. In three-dimensional co-culture, LAA promoted SplC infiltration and suppressed the formation of tumor spheres. Thus, LAA may exert antitumor effects through increased ROS production in cancer cells and effector T cell activation via increased energy metabolism. These results suggest that LAA, when used in combination with existing anti-cancer drugs, is likely to exhibit sensitizing effects in terms of both antitumor and antitumor immune effects, and future clinical studies are anticipated.
Collapse
Grants
- 23K16621 Ministry of Education, Culture, Sports, Science and Technology
- 19K16564 Ministry of Education, Culture, Sports, Science and Technology
- 23K10481 Ministry of Education, Culture, Sports, Science and Technology
- 21K11223 Ministry of Education, Culture, Sports, Science and Technology
- 22K16497 Ministry of Education, Culture, Sports, Science and Technology
- 21K06926 Ministry of Education, Culture, Sports, Science and Technology
- 20K21659 Ministry of Education, Culture, Sports, Science and Technology
Collapse
Affiliation(s)
- Shiori Mori
- Department of Molecular Pathology, School of Medicine, Nara Medical University, Kashihara 634-8521, Japan; (S.M.); (R.F.-T.); (R.O.); (H.O.); (K.F.); (Y.L.); (T.S.); (Y.N.); (S.K.)
- Department of Cancer Biology, Institute of Biomedical Science, Kansai Medical University, Osaka 573-1010, Japan
| | - Rina Fujiwara-Tani
- Department of Molecular Pathology, School of Medicine, Nara Medical University, Kashihara 634-8521, Japan; (S.M.); (R.F.-T.); (R.O.); (H.O.); (K.F.); (Y.L.); (T.S.); (Y.N.); (S.K.)
| | - Ruiko Ogata
- Department of Molecular Pathology, School of Medicine, Nara Medical University, Kashihara 634-8521, Japan; (S.M.); (R.F.-T.); (R.O.); (H.O.); (K.F.); (Y.L.); (T.S.); (Y.N.); (S.K.)
| | - Hitoshi Ohmori
- Department of Molecular Pathology, School of Medicine, Nara Medical University, Kashihara 634-8521, Japan; (S.M.); (R.F.-T.); (R.O.); (H.O.); (K.F.); (Y.L.); (T.S.); (Y.N.); (S.K.)
| | - Kiyomu Fujii
- Department of Molecular Pathology, School of Medicine, Nara Medical University, Kashihara 634-8521, Japan; (S.M.); (R.F.-T.); (R.O.); (H.O.); (K.F.); (Y.L.); (T.S.); (Y.N.); (S.K.)
| | - Yi Luo
- Department of Molecular Pathology, School of Medicine, Nara Medical University, Kashihara 634-8521, Japan; (S.M.); (R.F.-T.); (R.O.); (H.O.); (K.F.); (Y.L.); (T.S.); (Y.N.); (S.K.)
| | - Takamitsu Sasaki
- Department of Molecular Pathology, School of Medicine, Nara Medical University, Kashihara 634-8521, Japan; (S.M.); (R.F.-T.); (R.O.); (H.O.); (K.F.); (Y.L.); (T.S.); (Y.N.); (S.K.)
| | - Yukiko Nishiguchi
- Department of Molecular Pathology, School of Medicine, Nara Medical University, Kashihara 634-8521, Japan; (S.M.); (R.F.-T.); (R.O.); (H.O.); (K.F.); (Y.L.); (T.S.); (Y.N.); (S.K.)
| | - Ujjal Kumar Bhawal
- Research Institute of Oral Science, School of Dentistry at Matsudo, Nihon University, Matsudo 271-8587, Japan;
| | - Shingo Kishi
- Department of Molecular Pathology, School of Medicine, Nara Medical University, Kashihara 634-8521, Japan; (S.M.); (R.F.-T.); (R.O.); (H.O.); (K.F.); (Y.L.); (T.S.); (Y.N.); (S.K.)
- Department of Pathological Diagnosis, Nozaki Tokushukai Hospital, Daito 574-0074, Japan
| | - Hiroki Kuniyasu
- Department of Molecular Pathology, School of Medicine, Nara Medical University, Kashihara 634-8521, Japan; (S.M.); (R.F.-T.); (R.O.); (H.O.); (K.F.); (Y.L.); (T.S.); (Y.N.); (S.K.)
| |
Collapse
|
47
|
Wu H, Zhang W, Chang J, Wu J, Zhang X, Jia F, Li L, Liu M, Zhu J. Comprehensive analysis of mitochondrial-related gene signature for prognosis, tumor immune microenvironment evaluation, and candidate drug development in colon cancer. Sci Rep 2025; 15:6173. [PMID: 39979377 PMCID: PMC11842742 DOI: 10.1038/s41598-024-85035-2] [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: 12/29/2023] [Accepted: 12/30/2024] [Indexed: 02/22/2025] Open
Abstract
Colon adenocarcinoma (COAD), a common digestive system malignancy, involves crucial alterations in mitochondria-related genes influencing tumor growth, metastasis, and immune evasion. Despite limited studies on prognostic models for these genes in COAD, we established a mitochondrial-related risk prognostic model, including nine genes based on available TCGA and MitoCarta 3.0 databases, and validated its predictive power. We investigated the tumor microenvironment (TME), immune cell infiltration, complex cell communication, tumor mutation burden, and drug sensitivity of COAD patients using R language, CellChat, and additional bioinformatic tools from single-cell and bulk-tissue sequencing data. The risk model revealed significant differences in immune cell infiltration between high-risk and low-risk groups, with the strongest correlation found between tissue stem cells and macrophages in COAD. The risk score exhibited a robust correlation with TME signature genes and immune checkpoint molecules. Integrating the risk score with the immune score, microsatellite status, or TMB through TIDE analysis enhanced the accuracy of predicting immunotherapy benefits. Predicted drug efficacy offered options for both high- and low-risk group patients. Our study established a novel mitochondrial-related nine-gene prognostic signature, providing insights for prognostic assessment and clinical decision-making in COAD patients.
Collapse
Affiliation(s)
- Hao Wu
- Department of Medical Cell Biology and Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, 030001, China
| | - Wentao Zhang
- Department of Medical Cell Biology and Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, 030001, China
| | - Jingjia Chang
- Department of Medical Cell Biology and Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, 030001, China
| | - Jin Wu
- Department of Molecular & Cellular Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA
| | - Xintong Zhang
- Department of Medical Cell Biology and Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, 030001, China
| | - Fengfeng Jia
- Taiyuan Technology Transfer Promotion Center, Taiyuan, 030006, China
| | - Li Li
- Department of Medical Cell Biology and Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, 030001, China
| | - Ming Liu
- Department of Medical Cell Biology and Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, 030001, China.
| | - Jianjun Zhu
- Department of Medical Cell Biology and Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, 030001, China.
| |
Collapse
|
48
|
Hua D, Yang Q, Li X, Zhou X, Kang Y, Zhao Y, Wu D, Zhang Z, Li B, Wang X, Qi X, Chen Z, Cui G, Hong W. The combination of Clostridium butyricum and Akkermansia muciniphila mitigates DSS-induced colitis and attenuates colitis-associated tumorigenesis by modulating gut microbiota and reducing CD8 + T cells in mice. mSystems 2025; 10:e0156724. [PMID: 39840995 PMCID: PMC11834468 DOI: 10.1128/msystems.01567-24] [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: 12/05/2024] [Accepted: 12/23/2024] [Indexed: 01/23/2025] Open
Abstract
The gut microbiota is closely associated with inflammatory bowel disease (IBD) and colorectal cancer (CRC). Probiotics such as Clostridium butyricum (CB) or Akkermansia muciniphila (AKK) have the potential to treat inflammatory bowel disease (IBD) or colorectal cancer (CRC). However, research on the combined therapeutic effects and immunomodulatory mechanisms of CB and AKK in treating IBD or CRC has never been studied. This study evaluates the potential of co-administration of CB and AKK in treating DSS/AOM-induced IBD and colitis-associated CRC. Our results indicate that compared to mono-administration, the co-administration of CB and AKK not only significantly alleviates symptoms such as weight loss, colon shortening, and increased Disease Activity Index in IBD mice but also regulates the gut microbiota composition and effectively suppresses colonic inflammatory responses. In the colitis-associated CRC mice model, a combination of CB and AKK significantly alleviates weight loss and markedly reduces inflammatory infiltration of macrophages and cytotoxic T lymphocytes (CTLs) in the colon, thereby regulating anti-tumor immunity and inhibiting the occurrence of inflammation-induced CRC. In addition, we found that the combined probiotic therapy of CB and AKK can enhance the sensitivity of colitis-associated CRC mice to the immune checkpoint inhibitor anti-mouse PD-L1 (aPD-L1), significantly improving the anti-tumor efficacy of immunotherapy and the survival rate of colitis-associated CRC mice. Furthermore, fecal microbiota transplantation therapy showed that transplanting feces from CRC mice treated with the co-administration of CB and AKK into other CRC mice alleviated the tumor loads in the colon and significantly extended their survival rate. Our study suggests that the combined use of two probiotics, CB and AKK, can not only alleviate chronic intestinal inflammation but also inhibit the progression to CRC. This may be a natural and relatively safe method to support the gut microbiota and enhance the host's immunity against cancer. IMPORTANCE Our study suggests that the combined administration of CB and AKK probiotics, as opposed to a single probiotic strain, holds considerable promise in preventing the advancement of IBD to CRC. This synergistic effect is attributed to the ability of this probiotic combination to more effectively modulate the gut microbiota, curb inflammatory reactions, bolster the efficacy of immunotherapeutic approaches, and optimize treatment results via fecal microbiota transplantation.
Collapse
Affiliation(s)
- Dengxiong Hua
- Key Laboratory of Microbiology and Parasitology of Education Department of Guizhou, Guizhou Medical University, Guiyang, China
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & School/Hospital of Stomatology Guizhou Medical University, Guiyang, Guizhou, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry & Joint Laboratory of Helicobacter Pylori and Intestinal Microecology of Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control, Guiyang, Guizhou, China
| | - Qin Yang
- Key Laboratory of Microbiology and Parasitology of Education Department of Guizhou, Guizhou Medical University, Guiyang, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry & Joint Laboratory of Helicobacter Pylori and Intestinal Microecology of Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control, Guiyang, Guizhou, China
| | - Xiaowei Li
- Key Laboratory of Microbiology and Parasitology of Education Department of Guizhou, Guizhou Medical University, Guiyang, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry & Joint Laboratory of Helicobacter Pylori and Intestinal Microecology of Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control, Guiyang, Guizhou, China
| | - Xuexue Zhou
- Key Laboratory of Microbiology and Parasitology of Education Department of Guizhou, Guizhou Medical University, Guiyang, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry & Joint Laboratory of Helicobacter Pylori and Intestinal Microecology of Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control, Guiyang, Guizhou, China
| | - Yingqian Kang
- Key Laboratory of Microbiology and Parasitology of Education Department of Guizhou, Guizhou Medical University, Guiyang, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry & Joint Laboratory of Helicobacter Pylori and Intestinal Microecology of Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control, Guiyang, Guizhou, China
| | - Yan Zhao
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & School/Hospital of Stomatology Guizhou Medical University, Guiyang, Guizhou, China
| | - Daoyan Wu
- Key Laboratory of Microbiology and Parasitology of Education Department of Guizhou, Guizhou Medical University, Guiyang, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry & Joint Laboratory of Helicobacter Pylori and Intestinal Microecology of Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control, Guiyang, Guizhou, China
| | - Zhengrong Zhang
- Key Laboratory of Microbiology and Parasitology of Education Department of Guizhou, Guizhou Medical University, Guiyang, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry & Joint Laboratory of Helicobacter Pylori and Intestinal Microecology of Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control, Guiyang, Guizhou, China
| | - Boyan Li
- School of Public Health, Guizhou Medical University, Guiyang, Guizhou, China
| | - Xinxin Wang
- Key Laboratory of Microbiology and Parasitology of Education Department of Guizhou, Guizhou Medical University, Guiyang, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry & Joint Laboratory of Helicobacter Pylori and Intestinal Microecology of Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control, Guiyang, Guizhou, China
| | - Xiaolan Qi
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & School/Hospital of Stomatology Guizhou Medical University, Guiyang, Guizhou, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, Guiyang, Guizhou, China
| | - Zhenghong Chen
- Key Laboratory of Microbiology and Parasitology of Education Department of Guizhou, Guizhou Medical University, Guiyang, China
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & School/Hospital of Stomatology Guizhou Medical University, Guiyang, Guizhou, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry & Joint Laboratory of Helicobacter Pylori and Intestinal Microecology of Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control, Guiyang, Guizhou, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, Guiyang, Guizhou, China
| | - Guzhen Cui
- Key Laboratory of Microbiology and Parasitology of Education Department of Guizhou, Guizhou Medical University, Guiyang, China
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & School/Hospital of Stomatology Guizhou Medical University, Guiyang, Guizhou, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry & Joint Laboratory of Helicobacter Pylori and Intestinal Microecology of Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control, Guiyang, Guizhou, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, Guiyang, Guizhou, China
| | - Wei Hong
- Key Laboratory of Microbiology and Parasitology of Education Department of Guizhou, Guizhou Medical University, Guiyang, China
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education & School/Hospital of Stomatology Guizhou Medical University, Guiyang, Guizhou, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry & Joint Laboratory of Helicobacter Pylori and Intestinal Microecology of Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control, Guiyang, Guizhou, China
- Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, Guiyang, Guizhou, China
| |
Collapse
|
49
|
Ham H, Hirdler JB, Bihnam DT, Mao Z, Gicobi JK, Macedo BG, Rodriguez-Quevedo MF, Schultz DF, Correia C, Zhong J, Martinez KE, Banuelos A, Ashton DS, Lagnado AB, Guo R, Pessoa R, Pandey A, Li H, Lucien F, Borges da Silva H, Dong H, Billadeau DD. Lysosomal NKG7 restrains mTORC1 activity to promote CD8 + T cell durability and tumor control. Nat Commun 2025; 16:1628. [PMID: 39952956 PMCID: PMC11829009 DOI: 10.1038/s41467-025-56931-6] [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: 03/22/2024] [Accepted: 02/06/2025] [Indexed: 02/17/2025] Open
Abstract
During infection and cancer, mTORC1-mediated metabolic regulation impacts CD8+ T cell effector expansion and memory development. However, the mechanisms by which CD8+ T cells regulate mTORC1 to support their unique metabolic requirements remain unknown. Here we show that NKG7, a lysosomal protein whose expression is restricted to cytotoxic lymphocytes, negatively regulates mTORC1 recruitment and activation by inhibiting assembly and function of the lysosomal proton pump, vacuolar ATPase (v-ATPase). Human and mouse CD8+ T cells lacking NKG7 show more acidic lysosomes and increased activation of mTORC1 signaling, which could be reversed by inhibition of v-ATPase activity. In mice responding to LCMV infection, NKG7-deleted effector CD8+ T cells are less durable and generate fewer memory precursors, whereas induced expression of NKG7 in CD8+ T cells results in increased presence of intra-tumoral T cells. Overall, our work identifies NKG7 as a CD8+ T cell-specific regulator of mTORC1 activity, required for optimal immune responses.
Collapse
Affiliation(s)
- Hyoungjun Ham
- Division of Oncology Research, Mayo Clinic, Rochester, MN, USA.
- Department of Urology, Mayo Clinic, Rochester, MN, USA.
| | | | | | - Zhiming Mao
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | | - Cristina Correia
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Jun Zhong
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | | | - Alma Banuelos
- Department of Immunology, Mayo Clinic, Phoenix, AZ, USA
| | | | - Anthony B Lagnado
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Ruifeng Guo
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Jacksonville, FL, USA
| | | | - Akhilesh Pandey
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
- Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Hu Li
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | | | | | - Haidong Dong
- Department of Urology, Mayo Clinic, Rochester, MN, USA
- Department of Immunology, Mayo Clinic, Rochester, MN, USA
| | - Daniel D Billadeau
- Division of Oncology Research, Mayo Clinic, Rochester, MN, USA.
- Department of Immunology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
50
|
Yuan H, Jiao Y, Gao J, Wang T, Xia Y, Li K, Yang Y, Zhang J, Bao H, Wang L, Sun P, Li D, Li P, Cao Y, Zhao Z, Liu Z, Lu Z, Liu Y, Bai X. Enhancement of immune responses to classical swine fever virus E2 in mice by fusion or mixture with the porcine IL-28B. Appl Microbiol Biotechnol 2025; 109:44. [PMID: 39945936 PMCID: PMC11825588 DOI: 10.1007/s00253-024-13399-6] [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: 09/13/2024] [Revised: 12/23/2024] [Accepted: 12/28/2024] [Indexed: 02/16/2025]
Abstract
The E2 subunit vaccine has been considered a promising alternative to an attenuated classical swine fever (CSF) vaccine. However, it fails to induce a good cellular immune response. Given that immunogenic adjuvants can regulate the cellular immunity to achieve a maximum efficacy against antigens, immunostimulatory effects of porcine IL-28B on the CSF virus (CSFV) E2 subunit vaccine were evaluated in the present study. We expressed recombinant proteins E2-IL28B, E2, and IL-28B using CHO-S mammalian cells as an antigen expression platform, and three types of CSFV E2 subunit vaccines based on antigens E2-IL28B, E2 + IL-28B, and E2 were prepared, respectively. We found that both E2-IL28B and E2 + IL-28B antigens exhibited superior immunogenicity with dramatically induced antibody titers and neutralizing antibody levels than the E2 alone. Moreover, E2-IL28B or E2 + IL-28B, instead of E2, boosted cellular immune responses via obviously increasing the percentages of CD3+CD4+ T lymphocytes, promoting the lymphocyte proliferations, and enhancing the release of Th1-type cytokines. All results revealed that the inclusion of IL-28B, whether fused or mixed with E2, significantly elevated E2-induced immune potencies, suggesting that IL-28B could be used as a molecular adjuvant to optimize the design of E2 subunit vaccine for more effective controls of the CSF disease. KEY POINTS: • New CSF E2 subunit vaccine candidates were developed in which IL-28B was an immunoadjuvant • IL-28B significantly elevated the E2-induced immune potency whether it was fused or mixed with E2 • This study provided novel insights into the immunoregulatory properties of IL-28B used for the optimized subunit vaccine design.
Collapse
Affiliation(s)
- Hong Yuan
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Yunjuan Jiao
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Jie Gao
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Tao Wang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Yingju Xia
- National Reference Laboratory for Classical Swine Fever, China Institute of Veterinary Drug Control, Beijing, 100081, China
| | - Kun Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, 730046, China
| | - Yuxuan Yang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Jing Zhang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Huifang Bao
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Lihao Wang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Pu Sun
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Dong Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Pinghua Li
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Yimei Cao
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, 730046, China
| | - Zhixun Zhao
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Zaixin Liu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China
| | - Zengjun Lu
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China.
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, 730046, China.
| | - Yebing Liu
- National Reference Laboratory for Classical Swine Fever, China Institute of Veterinary Drug Control, Beijing, 100081, China.
| | - Xingwen Bai
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730000, China.
- Gansu Province Research Center for Basic Disciplines of Pathogen Biology, Lanzhou, 730046, China.
| |
Collapse
|