1
|
Jin H, Zhang D, Ma Y, Meng L, Huang S, Su H, Xu J, Yao Y. YTHDC2 manipulates anti-tumoral macrophage polarization and predicts favorable outcomes in triple negative breast cancer. NPJ Precis Oncol 2025; 9:119. [PMID: 40274959 DOI: 10.1038/s41698-025-00880-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 03/11/2025] [Indexed: 04/26/2025] Open
Abstract
Triple-negative breast cancer (TNBC) possesses high malignant and metastatic rates among all subtypes. Chemotherapy is a standard of care for TNBC but only a small moiety of patients achieved complete relief (CR) after chemotherapy. The recent concept of tumor ecosystem has provided new insights into solutions from an approach of enhancing anti-tumoral immunity of macrophages. We hereby observed a positive correlation of YTHDC2 abundance with anti-tumoral gene markers of macrophages. YTHDC2-high macrophages also exerted interactions with other immune cells such as T helper cells, cytotoxic T cells, and NK cells. Further investigation on the transcriptional regulatory network identified six transcriptional factors upregulated by YTHDC2, and they together influenced the expressions of TWISTNB and the oncogene MYC. Additionally, our survival analysis prompted that YTHDC2 is prognostic of higher chemo-therapeutic efficacy and better survival outcomes. We demonstrated that ample macrophage YTHDC2 indicates anti-tumoral phenotype polarization and propitious survival outcome in post-treatment TNBC patients (Clinical trial registry name: Chinese Clinical Trial Registry, Registration No.: ChiCTR2400084513, Registration Date: 2024-05-20).
Collapse
Affiliation(s)
- Hao Jin
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Dongbo Zhang
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Yufan Ma
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Lanlan Meng
- Shenshan Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Shanwei, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Cancer Pathogenesis and Precision Diagnosis and Treatment, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Shanwei, China
| | - Songyin Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Hongjun Su
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Jiannan Xu
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Yandan Yao
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China.
- Shenshan Medical Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Shanwei, Guangdong Province, China.
- Guangdong Provincial Key Laboratory of Cancer Pathogenesis and Precision Diagnosis and Treatment, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Shanwei, China.
| |
Collapse
|
2
|
Xi Y, Ma H, Liu X, Mu Q, An X, Li S, Liang H, Sun D, Ma R, Deng H, Wu Z, Zhang C, Liu G, Liu C. Epigenetically Reprogrammed Nanovesicles as Inverse Vaccines for Antigen-Specific Immune Tolerance in Autoimmune Diseases. NANO LETTERS 2025; 25:6725-6734. [PMID: 40213869 DOI: 10.1021/acs.nanolett.5c00986] [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: 04/24/2025]
Abstract
The development of antigen-specific immunotherapy for autoimmune diseases constitutes an important unmet clinical need. Here we present an innovative inverse vaccine platform leveraging epigenetic reprogramming to induce durable antigen-specific immune tolerance. This inverse vaccine (mDCNVreg) is constructed using artificial cell membrane nanovesicles derived from IFN-γ-primed regulatory dendritic cells subjected to epigenetic modulation. The engineered mDCNVreg features upregulated MHC-II expression enabling targeted antigen presentation, suppressed costimulatory molecules expression, and an enhanced coinhibitory molecules display. Through coordinated mechanisms involving enhanced lymphoid trafficking and phenotype stabilization, this platform significantly enhances antigen delivery to secondary lymphoid organs while maintaining tolerogenic potency. Crucially, mDCNVreg directly induces CD4+ T cell clonal anergy through epitope-specific interactions, establishing long-lasting immune tolerance. This work demonstrates a promising epigenetic engineering approach for reverse vaccine design in personalized autoimmune disease therapy.
Collapse
Affiliation(s)
- Yue Xi
- State Key Laboratory of Stress Biology and Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Huifeng Ma
- State Key Laboratory of Stress Biology and Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Xue Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Qianwen Mu
- State Key Laboratory of Stress Biology and Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Xiaoyu An
- State Key Laboratory of Stress Biology and Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Shuo Li
- State Key Laboratory of Stress Biology and Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Hao Liang
- State Key Laboratory of Stress Biology and Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Di Sun
- State Key Laboratory of Stress Biology and Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Rongrong Ma
- State Key Laboratory of Stress Biology and Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Haolan Deng
- State Key Laboratory of Stress Biology and Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Zhengyu Wu
- State Key Laboratory of Stress Biology and Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
| | - Chenhao Zhang
- State Key Laboratory of Stress Biology and Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Chao Liu
- State Key Laboratory of Stress Biology and Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
- Shenzhen Research Institute of Xiamen University, Shenzhen 518000, China
| |
Collapse
|
3
|
He L, Gan W, Xiao S, Shi Y, Fu M, Wu L, Zhang J. Association between systemic immune-inflammation index and chronic bronchitis: NHANES 2001-2018. Sci Rep 2025; 15:14113. [PMID: 40269079 PMCID: PMC12019561 DOI: 10.1038/s41598-025-97895-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 04/08/2025] [Indexed: 04/25/2025] Open
Abstract
The systemic immune-inflammation index (SII) is a newly identified marker of inflammation., and the relationship between chronic bronchitis (CB) and inflammation is closely associated. However, the influence of SII on CB remains unclear at present.This cross-sectional study was conducted using data from individuals with complete SII and CB records from the 2001-2018 National Health and Nutrition Examination Survey (NHANES). Binary weighted logistic regression was employed to investigate the relationship between SII and CB risk. Additionally, restricted cubic spline regression models and segmented regression models were used to examine nonlinear relationships and threshold effects. Receiver operating characteristic (ROC) curves were adopted to evaluate the predictive value of SII for CB. Stratified analysis was adopted to assess the association between SII and CB in different populations. After adjusting for all covariables, there was a significant positive relevance observed between log-transformed SII (log (SII)) with CB (OR = 1.52, 95% CI: 1.27-1.82, P < 0.001). A nonlinear dose-response relationship with the threshold of 8.14 was observed between log (SII) and CB risk. When log (SII) exceeded 8.14, each unit increase in log (SII) was associated with a 1.31-fold increase in the risk of CB (OR = 1.31, 95% CI: 1.22-1.40, P < 0.001). Furthermore, ROC curves revealed strong predictive capability of SII for CB (AUC = 0.729). Elevated SII levels are associated with an increased prevalence of CB. Furthermore, a non-linear association exists between SII and the increased risk of CB.
Collapse
Affiliation(s)
- Longxi He
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Weigang Gan
- Department of Otolaryngology, Head and Neck Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Sa Xiao
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yu Shi
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Mengjie Fu
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Li Wu
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jian Zhang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
4
|
Zhao S, Bai B, Zhu B, Cui Y, Deng W, Xie Z, Wang S, Wang X, Mao Y, Lu Y, Huang Y, You T, Sun W, Shen X, Lu X. Hyaluronic acid-modified doxorubicin-covalent organic framework nanoparticles triggered pyroptosis in combinations with immune checkpoint blockade for the treatment of breast cancer. Int J Biol Macromol 2025; 310:143265. [PMID: 40274165 DOI: 10.1016/j.ijbiomac.2025.143265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/03/2025] [Accepted: 04/15/2025] [Indexed: 04/26/2025]
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive malignancy, and the current treatment strategies have poor efficacy. Pyroptosis, a type of immunogenic cell death, significantly enhances antitumor immunity by triggering the release of numerous intracellular components. Here, we prepared a covalent-organic framework (COF) loaded with doxorubicin (DOX) and coated it with hyaluronic acid (HA) as a therapeutic delivery system (HA@DOX-COF) for the treatment of TNBC. Mechanistically, upon cellular uptake, HA@DOX-COF activated pyroptosis through the caspase-3/GSDME pathway. Moreover, HA@DOX-COF induced a pyroptosis-induced antitumor immune response and further augmented the efficacy of immune checkpoint blockade (ICB). Our results demonstrated that the combination of HA@DOX-COF and an anti-PD-1 antibody markedly inhibited tumor progression in a TNBC murine model. Overall, our work offers a potential approach for pyroptosis-induced immunotherapy for TNBC, which may increase the efficacy of ICB-based immunotherapy.
Collapse
Affiliation(s)
- Shengsheng Zhao
- Department of Gastrointestinal Surgery, Zhejiang International Scientific and Technological Cooperation Base of Translational Cancer Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Binglong Bai
- Department of Gastrointestinal Surgery, Zhejiang International Scientific and Technological Cooperation Base of Translational Cancer Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Bingzi Zhu
- Department of Gastrointestinal Surgery, Zhejiang International Scientific and Technological Cooperation Base of Translational Cancer Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Research Center of Basic Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yuekai Cui
- Department of Gastrointestinal Surgery, Zhejiang International Scientific and Technological Cooperation Base of Translational Cancer Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Research Center of Basic Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Wenhai Deng
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision, and Brain Health), School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Zuoliang Xie
- Department of Gastrointestinal Surgery, Zhejiang International Scientific and Technological Cooperation Base of Translational Cancer Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Research Center of Basic Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Shuaibin Wang
- Department of Gastrointestinal Surgery, Zhejiang International Scientific and Technological Cooperation Base of Translational Cancer Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Research Center of Basic Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Xiang Wang
- Department of Gastrointestinal Surgery, Zhejiang International Scientific and Technological Cooperation Base of Translational Cancer Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yiwen Mao
- Department of Breast Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yongyong Lu
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yingpeng Huang
- Department of Gastrointestinal Surgery, Zhejiang International Scientific and Technological Cooperation Base of Translational Cancer Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Tao You
- Department of Gastrointestinal Surgery, Zhejiang International Scientific and Technological Cooperation Base of Translational Cancer Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Weijian Sun
- Department of Gastrointestinal Surgery, Zhejiang International Scientific and Technological Cooperation Base of Translational Cancer Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| | - Xian Shen
- Department of Gastrointestinal Surgery, Zhejiang International Scientific and Technological Cooperation Base of Translational Cancer Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| | - Xufeng Lu
- Department of Gastrointestinal Surgery, Zhejiang International Scientific and Technological Cooperation Base of Translational Cancer Research, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Research Center of Basic Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| |
Collapse
|
5
|
Liu Y, Wang R, Han B, Zhu B, Ma Y, Yu Y, Bai L, Wei Q, Yang P. GPER1-mediated suppression of acute cellular rejection in murine cervical heart transplantation by estradiol. J Cardiothorac Surg 2025; 20:208. [PMID: 40247327 PMCID: PMC12004865 DOI: 10.1186/s13019-025-03432-8] [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: 09/11/2024] [Accepted: 04/06/2025] [Indexed: 04/19/2025] Open
Abstract
OBJECTIVE This study aimed to investigate the inhibitory effect of estradiol (E2) on acute cellular rejection following murine cervical heart transplantation and the role of G protein-coupled estrogen receptor 1 (GPER1). METHOD A murine cervical heart transplantation model was established and categorized into four groups: syngeneic group (C57BL/6 to C57BL/6), allogenic control group (BALB/c to C57BL/6), E2 group (BALB/c to C57BL/6), and E2 + G15 group (G15: one of GPER1 blocker) (BALB/c to C57BL/6). Survival time, pathological rejection grade, intimal hyperplasia area of coronary artery in donor heart, mRNA expression levels of TGF-β, IL-10, and Foxp3 in spleen, and protein expression levels of CD4 and CD25 in spleen membrane proteins, were compared among these groups. RESULTS The syngeneic group showed 100% survival, while survival in the allogenic control group was significantly reduced. E2 treatment prolonged survival compared to the allogenic control group, but survival decreased with the addition of E2 + G15. Rejection was reduced in the E2 group compared to the allogenic control group, whereas the E2 + G15 group exhibited increased rejection compared to the E2 group. Intimal hyperplasia was absent in the syngeneic group, significant in the allogenic control group, reduced in the E2 group compared to the allogenic control group, but greater in the E2 + G15 group compared to the E2 group. The expression of IL-10, Foxp3, and TGF-β in the allogenic control group was higher than in the syngeneic group. In the E2 group, IL-10, Foxp3, and CD25 were higher than in the allogenic control group, while TGF-β and CD4 expression were lower. The E2 + G15 group had lower IL-10, Foxp3, and CD25 expression but higher TGF-β expression than the E2 group. CONCLUSION Severe acute cellular rejection was observed following murine cervical heart transplantation. E2 suppresses acute rejection by enhancing Treg cell expression, mediated by GPER1.
Collapse
Affiliation(s)
- Yiwei Liu
- Department of Thoracic Surgery, Peking University Cancer Hospital Inner Mongolia Hospital & Inner Mongolia Medical University Affiliated Cancer Hospital, 42 Zhaowuda Road, Saihan District, Huhhot, Inner Mongolia Autonomous Region, 010020, China
- Department of Cardiothoracic Surgery, Jincheng People's Hospital, Jincheng, Shanxi, 048000, China
| | - Ran Wang
- Department of Cardiovascular Medicine, The First People's Hospital of Horqin District, Tongliao, Inner Mongolia Autonomous Region, 028000, China
| | - Bater Han
- Department of Thoracic Surgery, Peking University Cancer Hospital Inner Mongolia Hospital & Inner Mongolia Medical University Affiliated Cancer Hospital, 42 Zhaowuda Road, Saihan District, Huhhot, Inner Mongolia Autonomous Region, 010020, China
| | - Benben Zhu
- Department of Thoracic Surgery, Peking University Cancer Hospital Inner Mongolia Hospital & Inner Mongolia Medical University Affiliated Cancer Hospital, 42 Zhaowuda Road, Saihan District, Huhhot, Inner Mongolia Autonomous Region, 010020, China
| | - Yu Ma
- Department of Thoracic Surgery, Peking University Cancer Hospital Inner Mongolia Hospital & Inner Mongolia Medical University Affiliated Cancer Hospital, 42 Zhaowuda Road, Saihan District, Huhhot, Inner Mongolia Autonomous Region, 010020, China
| | - Yongjun Yu
- Department of Thoracic Surgery, Peking University Cancer Hospital Inner Mongolia Hospital & Inner Mongolia Medical University Affiliated Cancer Hospital, 42 Zhaowuda Road, Saihan District, Huhhot, Inner Mongolia Autonomous Region, 010020, China
| | - Lu Bai
- Department of Thoracic Surgery, Peking University Cancer Hospital Inner Mongolia Hospital & Inner Mongolia Medical University Affiliated Cancer Hospital, 42 Zhaowuda Road, Saihan District, Huhhot, Inner Mongolia Autonomous Region, 010020, China
| | - Qiyou Wei
- Department of Thoracic Surgery, Peking University Cancer Hospital Inner Mongolia Hospital & Inner Mongolia Medical University Affiliated Cancer Hospital, 42 Zhaowuda Road, Saihan District, Huhhot, Inner Mongolia Autonomous Region, 010020, China.
| | - Pengjie Yang
- Department of Thoracic Surgery, Peking University Cancer Hospital Inner Mongolia Hospital & Inner Mongolia Medical University Affiliated Cancer Hospital, 42 Zhaowuda Road, Saihan District, Huhhot, Inner Mongolia Autonomous Region, 010020, China.
| |
Collapse
|
6
|
Leng F, Merino-Urteaga R, Wang X, Zhang W, Ha T, Hur S. Ultrastable and versatile multimeric ensembles of FoxP3 on microsatellites. Mol Cell 2025; 85:1509-1524.e7. [PMID: 40179879 DOI: 10.1016/j.molcel.2025.03.005] [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: 11/21/2024] [Revised: 01/29/2025] [Accepted: 03/05/2025] [Indexed: 04/05/2025]
Abstract
Microsatellites are essential genomic components increasingly linked to transcriptional regulation. FoxP3, a transcription factor critical for regulatory T cell (Treg) development, recognizes TTTG repeat microsatellites by forming multimers along DNA. However, FoxP3 also binds a broader range of TnG repeats (n = 2-5), often at the edges of accessible chromatin regions. This raises questions about how FoxP3 adapts to sequence variability and the potential role of nucleosomes. Using cryoelectron microscopy and single-molecule analyses, we show that murine FoxP3 assembles into various distinct supramolecular structures, depending on DNA sequence. This structural plasticity enables FoxP3 to bridge 2-4 DNA duplexes, forming ultrastable structures that coordinate multiple genomic loci. Nucleosomes further facilitate FoxP3 assembly by inducing local DNA bending, creating a nucleus that recruits distal DNA elements through multiway bridging. Our findings thus reveal FoxP3's unusual ability to shapeshift to accommodate evolutionarily dynamic microsatellites and its potential to reinforce chromatin boundaries and three-dimensional genomic architecture.
Collapse
Affiliation(s)
- Fangwei Leng
- Howard Hughes Medical Institute and Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Institute of Immunology, Chinese Institutes for Medical Research, Beijing 100069, China
| | - Raquel Merino-Urteaga
- Howard Hughes Medical Institute and Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Department of Biology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Xi Wang
- Howard Hughes Medical Institute and Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Wenxiang Zhang
- Howard Hughes Medical Institute and Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Shanghai Institute of Immunology, Shanghai Jiaotong University, Shanghai 200025, China
| | - Taekjip Ha
- Howard Hughes Medical Institute and Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA.
| | - Sun Hur
- Howard Hughes Medical Institute and Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
7
|
Wang L, Wang S, Lin J, Li J, Wang M, Yu J, Sun J, Tang N, Jiao C, Ma J, Zhao X, Zhang H. Treg and intestinal myofibroblasts-derived Amphiregulin induced by TGF-β mediates intestinal fibrosis in Crohn's disease. J Transl Med 2025; 23:452. [PMID: 40247299 PMCID: PMC12004752 DOI: 10.1186/s12967-025-06413-6] [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: 01/23/2025] [Accepted: 03/23/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Intestinal fibrosis is a serious complication of Crohn's disease (CD), often resulting from chronic inflammation. However, the precise mechanisms through which inflammation induces intestinal fibrosis remain inadequately elucidated. METHODS A comprehensive single-cell atlas of full-thickness CD, provided by Dr. Florian Rieder, was subjected to reanalysis. Our study used a DSS-induced chronic colitis model in both wild-type (WT) and Areg-/- mice. Additionally, a CD45RBhi CD4+ T cell adoptive transfer model involving WT and Areg-/- Treg cells (Tregs) was used. The expressions of AREG in CD with or without intestinal fibrosis, Tregs and human intestinal myofibroblasts (MFs) were determined. The effect of AREG on proliferation/migration/activation in human intestinal MFs was determined. RESULTS Several types of cells were differentially expressed between stricture and non-stricture CD. Among T cells, Tregs accounted for a larger proportion and were significantly increased in stenotic tissues of stricture CD. Although DSS-induced colitis was more severe in Areg-/- mice, which developed less severe intestinal fibrosis compared with WT mice. The transfer of Areg-/- Tregs resulted in less severe fibrosis in Rag-/- mice than WT Tregs. Moreover, TGF-β stimulated AREG expression in Tregs and human intestinal MFs via activation of Smad3. CONCLUSION These findings demonstrated that AREG derived from Tregs and human intestinal MFs, induced by TGF-β, amplifies intestinal fibrotic reactions in experimental colitis as well as in human CD patients. Thus, the TGF-β-Smad3-AREG pathway could be a potential therapeutic target for treating fibrosis in CD.
Collapse
Affiliation(s)
- Lu Wang
- Department of Gastroenterology, First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, China
| | - Shu Wang
- Department of Gastroenterology, First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, China
| | - Junjie Lin
- Department of Gastroenterology, First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, China
| | - Jiajia Li
- Department of Gastroenterology, First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, China
| | - Mingyuan Wang
- Department of Gastroenterology, First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, China
| | - Jiang Yu
- Department of Gastroenterology, First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, China
| | - Junjian Sun
- Department of Gastroenterology, First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, China
| | - Nana Tang
- Department of Gastroenterology, First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, China
| | - Chunhua Jiao
- Department of Gastroenterology, First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, China
| | - Jingjing Ma
- Department of Gastroenterology, First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, China
| | - Xiaojing Zhao
- Department of Gastroenterology, First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, China.
| | - Hongjie Zhang
- Department of Gastroenterology, First Affiliated Hospital with Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
8
|
Lysandrou M, Kefala D, Vinnakota JM, Savvopoulos N, Zeiser R, Spyridonidis A. Regulatory T cell therapy for Graft-versus-Host Disease. Bone Marrow Transplant 2025:10.1038/s41409-025-02553-x. [PMID: 40240498 DOI: 10.1038/s41409-025-02553-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 02/17/2025] [Accepted: 03/11/2025] [Indexed: 04/18/2025]
Abstract
Graft-versus-Host Disease (GvHD) is the main cause of morbidity and mortality of allogeneic hematopoietic cell transplantation (allo-HCT). Conventional immunosuppressive pharmacotherapy remains the backbone of GvHD prevention and treatment with suboptimal outcomes especially for patients with refractory disease. Adoptive immunotherapy with regulatory T-cells (Treg) stands as an alternative approach that aims to restore immune tolerance and circumvent prolonged immunosuppression albeit preserving the beneficial Graft-versus-Leukaemia (GvL) effect. In this review, we summarise recent knowledge on Treg biology, clinical applications of various Tregs subtypes in the setting of GvHD and future endeavours of the field.
Collapse
Affiliation(s)
- Memnon Lysandrou
- Bone Marrow Transplantation Unit and Institute of Cell Therapy, University of Patras, Patras, Greece
- Department of Medicine I, Medical Center University of Freiburg, Faculty of Medicine, Albert Ludwigs University Freiburg, Freiburg, Germany
| | - Dionysia Kefala
- Bone Marrow Transplantation Unit and Institute of Cell Therapy, University of Patras, Patras, Greece
| | - Janaki Manoja Vinnakota
- Department of Medicine I, Medical Center University of Freiburg, Faculty of Medicine, Albert Ludwigs University Freiburg, Freiburg, Germany
| | - Nikolaos Savvopoulos
- Bone Marrow Transplantation Unit and Institute of Cell Therapy, University of Patras, Patras, Greece
| | - Robert Zeiser
- Department of Medicine I, Medical Center University of Freiburg, Faculty of Medicine, Albert Ludwigs University Freiburg, Freiburg, Germany
| | - Alexandros Spyridonidis
- Bone Marrow Transplantation Unit and Institute of Cell Therapy, University of Patras, Patras, Greece.
| |
Collapse
|
9
|
Huang C, Cheng EF, Ni J, Lyu Y. Baicalein prevents pregnancy loss by maintaining regulatory T cell activation through inhibition of the TLR4/NF-κB signaling pathway. Placenta 2025; 165:120-126. [PMID: 40245602 DOI: 10.1016/j.placenta.2025.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 04/10/2025] [Accepted: 04/13/2025] [Indexed: 04/19/2025]
Abstract
INTRODUCTION Unexplained recurrent spontaneous abortion (URSA) involves multifactorial etiologies, with regulatory T cells (Tregs) playing a pivotal role in maintaining immune tolerance during pregnancy. Baicalein, a flavonoid from Scutellaria baicalensis, exhibits anti-inflammatory and immunomodulatory properties. This study evaluates baicalein's therapeutic potential in mitigating URSA via the TLR4/NF-κB signaling pathway. METHODS A URSA mouse model was used, and baicalein was administered intraperitoneally. Pregnancy outcomes, abortion rates, and placental morphology were assessed on gestational day 14 (G14). Treg cells were quantified via flow cytometry, and gene/protein expression levels were analyzed by immunohistochemistry, Western blotting, and real-time PCR. In vitro experiments on ihESCs further investigated the role of the TLR4/NF-κB pathway. RESULTS Baicalein reduced abortion rates from 33.3 % in URSA mice to 21.6 % (low dose) and 14.8 % (high dose), improved embryonic development by altering placental structure and decidual morphology, and reduced inflammation at the maternal-fetal interface. It expanded Treg cells and enhanced the expression of IGFBP-1 and PRL, markers of endometrial decidualization, and decreased TLR4, p-P65, and P65 expression. In vitro, baicalein's effects were abrogated by TLR4 inhibition, confirming pathway specificity. DISCUSSION Baicalein improved pregnancy outcomes by enhancing Treg function and promoting decidualization via TLR4/NF-κB pathway inhibition. These findings highlight baicalein's potential as a therapeutic agent for URSA.
Collapse
Affiliation(s)
- Caiqun Huang
- Departments of Central Laboratory, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, 321000, China.
| | - E Fen Cheng
- Departments of Obstetrics and Gynecology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, 321000, China
| | - Jinping Ni
- School of Food Science and Technology, Jiangnan University, Wuxi, 214122, China
| | - Ying Lyu
- Departments of Obstetrics and Gynecology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, 321000, China
| |
Collapse
|
10
|
Qian X, Zai Z, Tao Y, Lv H, Hao M, Zhang L, Zhang X, Xu Y, Zhang Y, Chen F. Acidosis regulates immune progression in rheumatoid arthritis by promoting the expression of cytokines and co-stimulatory molecules in synovial fibroblasts. Mol Med 2025; 31:136. [PMID: 40234753 PMCID: PMC12001510 DOI: 10.1186/s10020-025-01181-x] [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: 01/14/2025] [Accepted: 03/24/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Tissue acidosis is a key characteristic of RA. It remains unclear whether acidosis promotes the formation of the complex adaptive immune landscape mainly characterized by T cell activation in RA by influencing synovial fibroblasts. This study aims to investigate the influence of acidosis on the immune microenvironment of RA by exploring the cytokine secretion and expression of co-stimulatory factors of RA synovial fibroblasts. METHODS The Bulk RNA-seq dataset (GSE89408, Normal = 23, RA = 150) was utilized for cytokine screening and the immune state assessment based on disease stage. RNA-seq was employed to investigate cytokine and co-stimulatory molecule expression following 6 h of acid stimulation, combined with Bulk RNA-seq data to evaluate contributions to RA. Human cytokine arrays were used to confirm cytokine accumulation in supernatants after 12 h of acid stimulation. Proteomics was applied to explore cellular functional states in RASFs under 6 h of acid stress, with joint RNA-seq analysis elucidating transcription factor activation. Validation of select high-throughput data was performed using qRT-PCR and immune-based assays. RESULTS Bulk RNA-seq and RNA-seq identified 56 differentially expressed cytokines at their intersection. Functional enrichment analysis demonstrated that acid stimulation enhanced cytokine secretion and T cell chemotaxis in RA synovial fibroblasts (RASFs). Cytokine array revealed that acid exposure increased the accumulation of growth factors (e.g., FGF, VEGF) by over twofold and promoted the expression of multiple inflammatory and chemotactic factors. Immune state analysis indicated that acid stimulation induced a complex immune landscape by upregulating co-stimulatory and antigen-presenting molecules. Proteomics showed that acid stress enhanced mitochondrial function and triggered metabolic reprogramming in RASFs. Integrated transcriptomic and proteomic analyses revealed that AP1 regulates gene expression in RASFs, with its activation further confirmed by Western blotting and immunofluorescence.
Collapse
Affiliation(s)
- Xuewen Qian
- School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- The Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - Zhuoyan Zai
- School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- The Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - Yuemin Tao
- School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- The Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - Huifang Lv
- School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- The Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - Mengjia Hao
- School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- The Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - Longbiao Zhang
- School of Pharmacy, Anhui Medical University, Hefei, 230032, China
- The Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, 230032, China
| | - Xiaoyue Zhang
- School of Public Health, Anhui Medical University, Hefei, 230032, China
| | - Yayun Xu
- Shenzhen Institute of Translational Medicine, Shenzhen Second People'S Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China.
| | - Yihao Zhang
- School of Public Health, Anhui Medical University, Hefei, 230032, China.
- Department of Health Inspection and Quarantine, School of Public Health, Anhui Medical University, Hefei, 230032, China.
| | - Feihu Chen
- School of Pharmacy, Anhui Medical University, Hefei, 230032, China.
- The Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
11
|
Meng X, Zhu Y, Liu K, Wang Y, Liu X, Liu C, Zeng Y, Wang S, Gao X, Shen X, Chen J, Tao S, Xu Q, Dong L, Shen L, Wang L. CXXC-finger protein 1 associates with FOXP3 to stabilize homeostasis and suppressive functions of regulatory T cells. eLife 2025; 13:RP103417. [PMID: 40183773 PMCID: PMC11970909 DOI: 10.7554/elife.103417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025] Open
Abstract
FOXP3-expressing regulatory T (Treg) cells play a pivotal role in maintaining immune homeostasis and tolerance, with their activation being crucial for preventing various inflammatory responses. However, the mechanisms governing the epigenetic program in Treg cells during their dynamic activation remain unclear. In this study, we demonstrate that CXXC-finger protein 1 (CXXC1) interacts with the transcription factor FOXP3 and facilitates the regulation of target genes by modulating H3K4me3 deposition. Cxxc1 deletion in Treg cells leads to severe inflammatory disease and spontaneous T cell activation, with impaired immunosuppressive function. As a transcriptional regulator, CXXC1 promotes the expression of key Treg functional markers under steady-state conditions, which are essential for the maintenance of Treg cell homeostasis and their suppressive functions. Epigenetically, CXXC1 binds to the genomic regulatory regions of Treg program genes in mouse Treg cells, overlapping with FOXP3-binding sites. Given its critical role in Treg cell homeostasis, CXXC1 presents itself as a promising therapeutic target for autoimmune diseases.
Collapse
Affiliation(s)
- Xiaoyu Meng
- Institute of Immunology and Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical CenterHangzhouChina
| | - Yezhang Zhu
- Department of Hematology, Tongji Hospital, School of Medicine, Tongji UniversityShanghaiChina
- Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center of Stem Cell Research, National Stem Cell Translational Resource Center, School of Life Sciences and Technology, Tongji UniversityShanghaiChina
| | - Kuai Liu
- Institute of Immunology and Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical CenterHangzhouChina
| | - Yuxi Wang
- Laboratory Animal Center, Zhejiang UniversityHangzhouChina
| | - Xiaoqian Liu
- Institute of Immunology and Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical CenterHangzhouChina
| | - Chenxin Liu
- Zhejiang University School of MedicineHangzhouChina
| | - Yan Zeng
- Institute of Immunology and Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical CenterHangzhouChina
| | - Shuai Wang
- Institute of Immunology and Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical CenterHangzhouChina
| | - Xianzhi Gao
- Institute of Immunology and Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical CenterHangzhouChina
| | - Xin Shen
- Co-Facility Center, Zhejiang University School of MedicineHangzhouChina
| | - Jing Chen
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
| | - Sijue Tao
- Laboratory Animal Center, Zhejiang UniversityHangzhouChina
| | - Qianying Xu
- Zhejiang University School of MedicineHangzhouChina
| | - Linjia Dong
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical CollegeHangzhouChina
| | - Li Shen
- MOE Key Laboratory of Biosystems Homeostasis & Protection and Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang UniversityHangzhouChina
- Department of Orthopedics Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang UniversityHangzhouChina
| | - Lie Wang
- Institute of Immunology and Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of MedicineHangzhouChina
- Zhejiang University School of MedicineHangzhouChina
- Liangzhu Laboratory, Zhejiang University Medical CenterHangzhouChina
- Laboratory Animal Center, Zhejiang UniversityHangzhouChina
- Future Health Laboratory, Innovation Center of Yangtze River Delta, Zhejiang UniversityJiaxingChina
| |
Collapse
|
12
|
Yang Y, Zhang C, Li H, He Q, Xie J, Liu H, Cui F, Lei Z, Qin X, Liu Y, Xu M, Huang S, Zhang X. A review of molecular interplay between inflammation and cancer: The role of lncRNAs in pathogenesis and therapeutic potential. Int J Biol Macromol 2025; 309:142824. [PMID: 40187457 DOI: 10.1016/j.ijbiomac.2025.142824] [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: 01/17/2025] [Revised: 03/31/2025] [Accepted: 04/02/2025] [Indexed: 04/07/2025]
Abstract
The inflammatory microenvironment (IME) has been demonstrated to facilitate the initiation and progression of tumors throughout the inflammatory process. Simultaneously, cancer can initiate or intensify the inflammatory response, thereby promoting tumor progression. This review examines the dual role of long non-coding RNAs (lncRNAs) in the interplay between inflammation and cancer. LncRNA modulate inflammation-induced cancer by influencing the activation of signaling pathways (NF-κB, Wnt/β-catenin, mTOR, etc), microRNA (miRNA) sponging, protein interactions, interactions with immune cells, and encoding short peptides. In contrast, lncRNAs also impact cancer-induced inflammatory processes by regulating cytokine expression, mediating tumor-derived extracellular vesicles (EVs), modulating intracellular reactive oxygen species (ROS) levels, and facilitating metabolic reprogramming. Furthermore, the therapeutic potential of lncRNA and the challenges of clinical translation were explicitly discussed as well. Overall, this review aims to provide a comprehensive and systematic resource for future researchers investigating the impact of lncRNAs on inflammation and cancer.
Collapse
Affiliation(s)
- Yan Yang
- Department of Pharmacy, The Third People's Hospital of Chengdu, Chengdu, China; School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Chuxi Zhang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Huacui Li
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China; Tangshan Institute of Southwest Jiaotong University, Tangshan, China
| | - Qin He
- Department of Pharmacy, The Third People's Hospital of Chengdu, Chengdu, China
| | - Jiang Xie
- Department of Pediatrics, The Third People's Hospital of Chengdu, Chengdu, China
| | - Hongmei Liu
- Department of Pharmacy, The Third People's Hospital of Chengdu, Chengdu, China
| | - Fenfang Cui
- Department of Pharmacy, The Third People's Hospital of Chengdu, Chengdu, China
| | - Ziqin Lei
- Department of Pharmacy, The Third People's Hospital of Chengdu, Chengdu, China
| | - Xiaoyan Qin
- Department of Pharmacy, The Third People's Hospital of Chengdu, Chengdu, China
| | - Ying Liu
- Department of Pharmacy, The Third People's Hospital of Chengdu, Chengdu, China
| | - Min Xu
- Department of Pharmacy, The Third People's Hospital of Chengdu, Chengdu, China.
| | - Shuai Huang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China.
| | - Xu Zhang
- Department of Pharmacy, Chengdu Integrated TCM & Western Medicine Hospital, Chengdu University of TCM, Chengdu, China.
| |
Collapse
|
13
|
Luo J, Chen K, Nong X. Potential regulation of artesunate on bone metabolism through suppressing inflammatory infiltration in type 2 diabetes mellitus. Immunopharmacol Immunotoxicol 2025; 47:147-158. [PMID: 39762719 DOI: 10.1080/08923973.2024.2444953] [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: 01/24/2024] [Accepted: 12/15/2024] [Indexed: 03/29/2025]
Abstract
OBJECTIVE Osteoimmunology is an emerging field that explores the interplay between bone and the immune system. The immune system plays a critical role in the pathogenesis of diabetes and significantly affects bone homeostasis. Artesunate, a first-line treatment for malaria, is known for its low toxicity and multifunctional properties. Increasing evidence suggests that artesunate possesses anti-inflammatory, immunoregulatory, and osteogenic effects. This review aims to explore the relationship between immune regulation and bone metabolism in type 2 diabetes (T2DM) and to investigate the potential therapeutic application of artesunate. METHODS This review systematically examines literature from PubMed/Medline, Elsevier, Web of Science, Embase, the International Diabetes Federation, and other relevant databases. RESULTS This review synthesizes evidence from multiple sources to delineate the relationship between T lymphocytes and T2DM, the regulation of T lymphocyte subsets in bone metabolism, and the effects of artesunate on both T lymphocytes and bone metabolism. Recent studies suggest a bidirectional regulatory relationship between T2DM and T lymphocytes (CD4+ T and CD8+ T) during the onset and progression of the disease, with inflammatory and anti-inflammatory cytokines serving as key mediators. T lymphocyte subsets and their cytokines play a pivotal role in regulating osteogenesis and osteoclastogenesis in pathological conditions. Furthermore, artesunate has shown promise in modulating inflammatory infiltration and bone metabolism. CONCLUSION The accumulated evidence indicates that artesunate exerts regulatory effects on bone metabolism in T2DM by influencing T lymphocyte differentiation.
Collapse
Affiliation(s)
- Jinghong Luo
- Department of Oral & Maxillofacial Surgery, College of Stomatology, Guangxi Medical University, Nanning, Guangxi, China
| | - Kun Chen
- Department of Oral & Maxillofacial Surgery, College of Stomatology, Guangxi Medical University, Nanning, Guangxi, China
| | - Xiaolin Nong
- Department of Oral & Maxillofacial Surgery, College of Stomatology, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
14
|
Wu G, Su T, Zhou P, Tang R, Zhu X, Wang J, Chao M, Fan L, Yan H, Ye P, Yu D, Gao F, Chen H. Engineering M2 macrophage-derived exosomes modulate activated T cell cuproptosis to promote immune tolerance in rheumatoid arthritis. Biomaterials 2025; 315:122943. [PMID: 39509857 DOI: 10.1016/j.biomaterials.2024.122943] [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: 09/11/2024] [Revised: 10/31/2024] [Accepted: 11/01/2024] [Indexed: 11/15/2024]
Abstract
Nanomedicines for immune modulation have made advancements in the treatment of rheumatoid arthritis (RA). However, due to aberrations in patients' immune systems, inducing antigen-specific immune tolerance while halting disease progression remains a significant challenge. Here, we develop a highly targeted multifunctional nanocomplex, termed M2Exo@CuS-CitP-Rapa (M2CPR), with the aim of selectively inhibiting inflammatory immune reactions while promoting immune tolerance towards specific antigens. M2CPR specifically targets inflammatory tissues in RA, delivering CuS NPs, CitP, Rapa, and endogenous anti-inflammatory factors, thereby ameliorating the inflammatory joint microenvironment. CuS NPs induce Cuproptosis of activated T cells, whose fragments are engulfed by resident or recruited macrophages, resulting in abundant production of TGF-β. TGF-β acts synergistically with Rapa to induce the iDCs into tDCs. tDCs present CitP to Naive T cells, promoting Tregs differentiation. Tregs, in turn, produce more TGF-β, inducing tDCs differentiation, thereby establishing a cycle of immune tolerance. Through in vitro and in vivo experiments, we validate that M2CPR can induce robust and durable antigen-specific immune tolerance, offering a new paradigm for RA therapy.
Collapse
Affiliation(s)
- Guoquan Wu
- Department of Orthopedics, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China; Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Tianyu Su
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Peng Zhou
- Department of Orthopedics, The Affiliated Huai'an Hospital of Xuzhou Medical University, The Second People's Hospital of Huai'an, Huai'an, 223002, China
| | - Rongze Tang
- Department of Orthopedics, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China; Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Xu Zhu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Jin Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Minghao Chao
- Department of Orthopedics, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China; Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Liying Fan
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Hanrong Yan
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Peng Ye
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Dehong Yu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Fenglei Gao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China.
| | - Hongliang Chen
- Department of Orthopedics, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China.
| |
Collapse
|
15
|
Prasongtanakij S, Soontrapa K, Thumkeo D. The role of prostanoids in regulatory T cells and their implications in inflammatory diseases and cancers. Eur J Cell Biol 2025; 104:151482. [PMID: 40184828 DOI: 10.1016/j.ejcb.2025.151482] [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: 12/06/2024] [Revised: 03/05/2025] [Accepted: 03/11/2025] [Indexed: 04/07/2025] Open
Abstract
Regulatory T cells (Tregs) play an important role in the immune system through the regulation of immunological self-tolerance and homeostasis. Furthermore, increasing evidence suggests the potential contribution of Tregs beyond immunity in the process of repairing various injured tissues. Tregs are generally characterised by the constitutive expression of forkhead box protein 3 (FOXP3) transcription factor in the nucleus and high expression levels of CD25 and CTLA-4 on the cell surface. To date, a large number of molecules have been identified as key regulators of Treg differentiation and function. Among these molecules are prostanoids, which are multifaceted lipid mediators. Prostanoids are produced from arachidonic acid through the catalytic activity of the enzyme cyclooxygenase and exert their functions through the 9 cognate receptors, DP1-2, EP1-EP4, FP, IP and TP. We briefly review previous studies on the regulatory mechanism of Tregs and then discuss recent works on the modulatory role of prostanoids.
Collapse
Affiliation(s)
- Somsak Prasongtanakij
- Laboratory of Immunopharmacology, Kyoto University Graduate School of Medicine, Japan
| | - Kitipong Soontrapa
- Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Thailand
| | - Dean Thumkeo
- Laboratory of Immunopharmacology, Kyoto University Graduate School of Medicine, Japan; Center for Medical Education and Internationalization, Kyoto University Faculty of Medicine, Japan.
| |
Collapse
|
16
|
Xu L, Shen T, Li Y, Wu X. The Role of M 6A Modification in Autoimmunity: Emerging Mechanisms and Therapeutic Implications. Clin Rev Allergy Immunol 2025; 68:29. [PMID: 40085180 DOI: 10.1007/s12016-025-09041-6] [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] [Accepted: 03/03/2025] [Indexed: 03/16/2025]
Abstract
N6-methyladenosine (m6A), a prevalent and essential RNA modification, serves a key function in driving autoimmune disease pathogenesis. By modulating immune cell development, activation, migration, and polarization, as well as inflammatory pathways, m6A is crucial in forming innate defenses and adaptive immunity. This article provides a comprehensive overview of m6A modification features and reveals how its dysregulation affects the intensity and persistence of immune responses, disrupts immune tolerance, exacerbates tissue damage, and promotes the development of autoimmunity. Specific examples include its contributions to systemic autoimmune disorders like lupus and rheumatoid arthritis, as well as conditions that targeting specific organs like multiple sclerosis and type 1 diabetes. Furthermore, this review explores the therapeutic promise of target m6A-related enzymes ("writers," "erasers," and "readers") and summarizes recent advances in intervention strategies. By focusing on the mechanistic and therapeutic implications of m6A modification, this review sheds light on its role as a promising tool for both diagnosis and treatment in autoimmune disorders, laying the foundation for advancements in customized medicine.
Collapse
Affiliation(s)
- Liyun Xu
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Tian Shen
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Yongzhen Li
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| | - Xiaochuan Wu
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
17
|
Huang Y, Liu P, Xu Y, Qian C, Wu T, Li T. Plasma Exosomes Derived from Patients with Primary Immune Thrombocytopenia Attenuate TBX21 + Regulatory T Cell-Mediated Immune Suppression via MiR-363-3p. Inflammation 2025:10.1007/s10753-025-02275-8. [PMID: 40032779 DOI: 10.1007/s10753-025-02275-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/14/2025] [Accepted: 02/18/2025] [Indexed: 03/05/2025]
Abstract
Primary Immune Thrombocytopenia (ITP) is characterized by reduced immunosuppressive function of regulatory T cells (Tregs), contributing to immune imbalance and decreased platelet counts. However, the mechanisms behind this reduced efficacy of Tregs remain unclear. Our study used a variety of methods, including Treg function assays, cytokine analysis, and single-cell sequencing, to explore these mechanisms. We found that exosomes from ITP patients inhibited TBX21 expression in Tregs, and impaired their ability to suppress Th1 cells. At the single-cell level, Tregs with high TBX21 expression were identified, and the activity of the TBX21 regulon was found to be enhanced in early-stage Treg subpopulations. We also discovered that ARID3A interacted with SPI1 and TBX21 gene regions, indicating a regulatory relationship between ARID3A, SPI1, and TBX21. Additionally, exosomes in ITP patients' plasma contained elevated levels of miR-363-3p, which negatively correlated with platelet count. These exosomes transferred miR-363-3p to Tregs, downregulating ARID3A, SPI1, and TBX21 expression, thereby weakening Tregs' ability to suppress conventional CD4 + T cells. In conclusion, exosomes from ITP patients reduced Treg function through the ARID3A/SPI1/TBX21 axis by miR-363-3p, diminishing their ability to regulate Th1 cells and contributing to the immune dysfunction observed in ITP.
Collapse
Affiliation(s)
- Yuanlan Huang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
- Department of Blood Transfusion, Naval Specialty Medical Center, Naval Medical University, Shanghai, 200000, China
| | - Peng Liu
- Department of Blood Transfusion, No.971 Hospital of the PLA Navy, Qingdao, China
| | - Ying Xu
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Cheng Qian
- Department of Laboratory Medicine, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Tianqin Wu
- Suzhou100 Hospital, Suzhou, 215006, China
| | - Tengda Li
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| |
Collapse
|
18
|
Gu C, Liu Y, Lv J, Zhang C, Huang Z, Jiang Q, Gao Y, Tao T, Su Y, Chen B, Jia R, Liu X, Su W. Kurarinone regulates Th17/Treg balance and ameliorates autoimmune uveitis via Rac1 inhibition. J Adv Res 2025; 69:381-398. [PMID: 38522752 PMCID: PMC11954799 DOI: 10.1016/j.jare.2024.03.013] [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: 10/26/2023] [Revised: 03/14/2024] [Accepted: 03/17/2024] [Indexed: 03/26/2024] Open
Abstract
INTRODUCTION Autoimmune uveitis (AU) is a severe intraocular autoimmune disorder with a chronic disease course and a high rate of blindness. Kurarinone (KU), a major component of the traditional Chinese medicine Sophorae Flavescentis Radix, possesses a wide spectrum of activities and has been used to treat several inflammation-related diseases. OBJECTIVE We aimed to investigate the effects of KU on AU and its modulatory mechanisms. METHODS We used an experimental autoimmune uveitis (EAU) animal model and characterized the comprehensive immune landscape of KU-treated EAU mice using single-cell RNA sequencing (scRNA-seq). The retina and lymph nodes were analyzed. The siRNAs and selective inhibitors were used to study the signaling pathway. The effect of KU on peripheral blood mononuclear cells (PBMCs) from uveitis patients was also examined. RESULTS We found that KU relieved chorioretinal lesions and immune cell infiltration in EAU model mice. Subsequent single-cell analysis revealed that KU downregulated the EAU-upregulated expression of inflammatory and autoimmune-related genes and suppressed pathways associated with immune cell differentiation, activation, and migration in a cell-specific manner. KU was implicated in restoring T helper 17 (Th17)/regulatory T (Treg) cell balance by alleviating inflammatory injury and elevating the expression of modulatory mediators in Tregs, while simultaneously ameliorating excessive inflammation by Th17 cells. Furthermore, Rac1 and the Id2/Pim1 axis potentiated the pathogenicity of Th17 cells during EAU, which was inhibited by KU treatment, contributing to the amelioration of EAU-induced inflammation and treatment of AU. In addition, KU suppressed inflammatory cytokine production in activated human PBMCs by inhibiting Rac1. Integration of the glucocorticoid-treated transcriptome suggests that KU has immunomodulatory effects on lymphocytes. CONCLUSION Our study constructed a high-resolution atlas of the immunoregulatory effects of KU treatment on EAU and identified its potential therapeutic mechanisms, which hold great promise in treating autoimmune disorders.
Collapse
Affiliation(s)
- Chenyang Gu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Yidan Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Jianjie Lv
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Chun Zhang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhaohao Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Qi Jiang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Yuehan Gao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Tianyu Tao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Yuhan Su
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China; Department of Clinical Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou 510060, China
| | - Binyao Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Renbing Jia
- Department of Ophthalmology, Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai 200011, China
| | - Xiuxing Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China.
| | - Wenru Su
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China.
| |
Collapse
|
19
|
Hernández-Bello J, Preciado-Aguiar MS, Muñoz-Valle JF, Baños-Hernández CJ, García-Arellano S, Alvarado-Navarro A. Influence of FOXP3 rs2280883 and rs3761548 Variants on IL-10 and TGF-β1 Serum Levels and Plaque Psoriasis Risk in the Mexican Population. Int J Mol Sci 2025; 26:1789. [PMID: 40076417 PMCID: PMC11898888 DOI: 10.3390/ijms26051789] [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: 01/25/2025] [Revised: 02/14/2025] [Accepted: 02/18/2025] [Indexed: 03/14/2025] Open
Abstract
Plaque psoriasis (PP) is a chronic immune-mediated skin disorder with a genetic basis, characterized by abnormal T-cell responses. This study investigated the role of FOXP3 gene variants rs2280883 and rs3761548 in T-cell regulation through their effects on IL-10 and TGF-β1 cytokine levels and their association with PP risk. A case-control study was conducted, including 101 individuals with PP and 106 healthy controls from the Mexican population. Genotyping of FOXP3 variants was performed using PCR-RFLP, and cytokine levels were measured with ELISA kits. Significant differences in allele and genotype frequencies of the rs2280883 variant were observed between PP patients and controls, suggesting an association with an increased risk of PP. IL-10 levels were found to be elevated in PP patients, regardless of FOXP3 gene variants, indicating that cytokine dysregulation in PP may involve alternative pathways independent of FOXP3-mediated regulatory T-cell (Treg) function. No significant differences were detected in TGF-β1 levels or rs3761548 genotype frequencies across the study groups. In conclusion, the rs2280883 variant in the FOXP3 gene is significantly associated with a higher risk of developing PP in the Mexican population, while dysregulated IL-10 levels suggest a complex cytokine interaction beyond Treg activity.
Collapse
Affiliation(s)
- Jorge Hernández-Bello
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico; (J.H.-B.); (J.F.M.-V.); (C.J.B.-H.); (S.G.-A.)
| | - Miriam Sarahi Preciado-Aguiar
- Centro de Investigación en Inmunología y Dermatología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico;
| | - José Francisco Muñoz-Valle
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico; (J.H.-B.); (J.F.M.-V.); (C.J.B.-H.); (S.G.-A.)
| | - Christian Johana Baños-Hernández
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico; (J.H.-B.); (J.F.M.-V.); (C.J.B.-H.); (S.G.-A.)
| | - Samuel García-Arellano
- Instituto de Investigación en Ciencias Biomédicas, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico; (J.H.-B.); (J.F.M.-V.); (C.J.B.-H.); (S.G.-A.)
| | - Anabell Alvarado-Navarro
- Centro de Investigación en Inmunología y Dermatología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico;
| |
Collapse
|
20
|
Muhs T, Ljubojevic-Holzer S, Sattler S. Anti-inflammatory Therapies for Ischemic Heart Disease. Curr Cardiol Rep 2025; 27:57. [PMID: 39969632 PMCID: PMC11839821 DOI: 10.1007/s11886-025-02211-0] [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] [Accepted: 01/31/2025] [Indexed: 02/20/2025]
Abstract
PURPOSE OF REVIEW The inclusion of immunomodulatory strategies as supportive therapies in ischemic heart disease (IHD) has garnered significant support over recent years. Several such approaches appear to be unified through their ultimate target, the NLRP3 inflammasome. This review presents a brief update on immunomodulatory strategies in the continuum of conditions constituting ischemic heart disease and emphasising on the seemingly unifying mechanism of NLRP3 activation as well as modulation across these conditions. RECENT FINDINGS The NLRP3 inflammasome is a multiprotein complex assembled upon inflammatory stimulation, causing the release of pro-inflammatory cytokines and initiating pyroptosis. The NLRP3 pathway is relevant in inflammatory signalling of cardiac immune cells as well as non-immune cells in the myocardium, including cardiomyocytes, fibroblasts and endothelial cells. In addition to a focus on clinical outcome and efficacy trials of targeting NLRP3-related pathways, the potential connection between immunomodulation in cardiology and the NLRP3 pathway is currently being explored in preclinical trials. Colchicine, cytokine-based approaches and SGLT2 inhibitors have emerged as promising agents. However, the conditions comprising IHD including atherosclerosis, coronary artery disease (CAD), myocardial infarction (MI) and ischemic cardiomyopathy/heart failure (iCMP/HF) are not equally amenable to immunomodulation with the respective drugs. Atherosclerosis, coronary artery disease and ischemic cardiomyopathy are affected by chronic inflammation, but the immunomodulatory approach to acute inflammation in the post-MI setting remains a pharmacological challenge, as detrimental and regenerative effects of myocardial inflammation are initiated in unison. The NLRP3 inflammasome lies at the center of cell mediated inflammation in IHD. Recent trial evidence has highlighted anti-inflammatory effects of colchicine, interleukin-based therapy as well as SGLT2i in IHD and that the respective drugs modulate the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Tillmann Muhs
- Department of Pharmacology, Otto-Loewi Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, 8010, Graz, Austria
| | - Senka Ljubojevic-Holzer
- Department of Cardiology, LKH Univ. Klinikum Graz, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria
| | - Susanne Sattler
- Department of Pharmacology, Otto-Loewi Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, 8010, Graz, Austria.
- Department of Cardiology, LKH Univ. Klinikum Graz, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria.
- National Heart and Lung Institute, Imperial College London, Hammersmith Campus, Du Cane Road, London, W12 0NN, UK.
| |
Collapse
|
21
|
An Q, Duan L, Wang Y, Wang F, Liu X, Liu C, Hu Q. Role of CD4 + T cells in cancer immunity: a single-cell sequencing exploration of tumor microenvironment. J Transl Med 2025; 23:179. [PMID: 39953548 PMCID: PMC11829416 DOI: 10.1186/s12967-025-06167-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 01/22/2025] [Indexed: 02/17/2025] Open
Abstract
Recent oncological research has intensely focused on the tumor immune microenvironment (TME), particularly the functions of CD4 + T lymphocytes. CD4+ T lymphocytes have been implicated in antigen presentation, cytokine release, and cytotoxicity, suggesting their contribution to the dynamics of the TME. Furthermore, the application of single-cell sequencing has yielded profound insights into the phenotypic diversity and functional specificity of CD4+ T cells in the TME. In this review, we discuss the current findings from single-cell analyses, emphasizing the heterogeneity of CD4+ T cell subsets and their implications in tumor immunology. In addition, we review the critical signaling pathways and molecular networks underpinning CD4+ T cell activities, thereby offering novel perspectives on therapeutic targets and strategies for cancer treatment and prognosis.
Collapse
Affiliation(s)
- Qi An
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Li Duan
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yuanyuan Wang
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Fuxin Wang
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xiang Liu
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Gannan Medical University, Jiangxi, 341000, China.
| | - Chao Liu
- Department of Radiation Oncology, Peking University First Hospital, Beijing, 100034, China.
| | - Qinyong Hu
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
22
|
Yuan L, Jiang N, Li Y, Wang X, Wang W. RGS1 Enhancer RNA Promotes Gene Transcription by Recruiting Transcription Factor FOXJ3 and Facilitates Osteoclastogenesis Through PLC-IP3R-dependent Ca 2+ Response in Rheumatoid Arthritis. Inflammation 2025; 48:447-463. [PMID: 38904871 DOI: 10.1007/s10753-024-02067-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 06/22/2024]
Abstract
Recent evidence has highlighted the functions of enhancers in modulating transcriptional machinery and affecting the development of human diseases including rheumatoid arthritis (RA). Enhancer RNAs (eRNAs) are RNA molecules transcribed from active enhancer regions. This study investigates the specific function of eRNA in gene transcription and osteoclastogenesis in RA. Regulator of G protein signaling 1 (RGS1)-associated eRNA was highly activated in osteoclasts according to bioinformatics prediction. RGS1 mRNA was increased in mice with collagen-induced arthritis as well as in M-CSF/soluble RANKL-stimulated macrophages (derived from monocytes). This was ascribed to increased RGS1 eRNA activity. Silencing of 5'-eRNA blocked the binding between forkhead box J3 (FOXJ3) and the RGS1 promoter, thus suppressing RGS1 transcription. RGS1 accelerated osteoclastogenesis through PLC-IP3R-dependent Ca2+ response. Knockdown of either FOXJ3 or RGS1 ameliorated arthritis severity, improved pathological changes, and reduced osteoclastogenesis and bone erosion in vivo and in vitro. However, the effects of FOXJ3 silencing were negated by RGS1 overexpression. In conclusion, this study demonstrates that the RGS1 eRNA-driven transcriptional activation of the FOXJ3/RGS1 axis accelerates osteoclastogenesis through PLC-IP3R dependent Ca2+ response in RA. The finding may offer novel insights into the role of eRNA in gene transcription and osteoclastogenesis in RA.
Collapse
MESH Headings
- Animals
- Arthritis, Rheumatoid/metabolism
- Arthritis, Rheumatoid/genetics
- Arthritis, Rheumatoid/pathology
- RGS Proteins/genetics
- RGS Proteins/metabolism
- Mice
- Osteoclasts/metabolism
- Osteoclasts/pathology
- Osteogenesis/genetics
- Osteogenesis/physiology
- Inositol 1,4,5-Trisphosphate Receptors/metabolism
- Forkhead Transcription Factors/metabolism
- Forkhead Transcription Factors/genetics
- Humans
- Transcription, Genetic
- Arthritis, Experimental/metabolism
- Arthritis, Experimental/genetics
- Arthritis, Experimental/pathology
- Enhancer Elements, Genetic
- Calcium/metabolism
- Enhancer RNAs
Collapse
Affiliation(s)
- Lin Yuan
- Department of Health Management, The First Affiliated Hospital of China Medical University, No. 155, Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, P.R. of China
| | - Nan Jiang
- Department of Price, The First Affiliated Hospital of China Medical University, Shenyang, 110001, Liaoning, P.R. China
| | - Yuxuan Li
- Department of Rheumatology and Immunology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, P.R. China
| | - Xin Wang
- Department of Health Management, The First Affiliated Hospital of China Medical University, No. 155, Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, P.R. of China
| | - Wei Wang
- Department of Health Management, The First Affiliated Hospital of China Medical University, No. 155, Nanjing North Street, Heping District, Shenyang, 110001, Liaoning, P.R. of China.
| |
Collapse
|
23
|
Kim J, Li J, Wei J, Lim SA. Regulatory T Cell Metabolism: A Promising Therapeutic Target for Cancer Treatment? Immune Netw 2025; 25:e13. [PMID: 40078783 PMCID: PMC11896657 DOI: 10.4110/in.2025.25.e13] [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: 12/30/2024] [Revised: 02/09/2025] [Accepted: 02/11/2025] [Indexed: 03/14/2025] Open
Abstract
Regulatory T (Treg) cells are essential for maintaining immune homeostasis by suppressing excessive immune responses. In the context of cancer, however, Tregs promote immune evasion and tumor progression, particularly through their unique adaptations within the tumor microenvironment (TME). Recent research has emphasized how metabolic characteristics shape Treg activation, migration, and immunosuppressive function, revealing the impact of metabolic pathways on Treg fitness in homeostasis and within the TME. In this review, we first provide an overview of Tregs in cancer immunology, discussing their immunosuppressive roles and properties specific to the TME. We then examine the metabolic requirements for Treg activation and migration under normal conditions, followed by a discussion of how hypoxia, lactate accumulation, nutrient limitation, oxidative stress, and other TME-specific factors alter Treg metabolism and contribute to cancer immune evasion. Finally, we explore therapeutic strategies that target Treg metabolism within the TME, including pharmacological modulation of specific metabolic pathways to diminish Treg-mediated immunosuppression. Thus, we could suggest future directions and clinical implications for Treg-targeted metabolic modulation as a complementary approach in cancer treatment, setting the stage for novel strategies in immunotherapy.
Collapse
Affiliation(s)
- Jihyoun Kim
- Department of Life Science, Ewha Womans University, Seoul 03760, Korea
| | - Jiaoran Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Jun Wei
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Seon Ah Lim
- Department of Life Science, Ewha Womans University, Seoul 03760, Korea
- Research Center for Cellular Homeostasis, Ewha Womans University, Seoul 03760, Korea
- Multitasking Macrophage Research Center, Ewha Womans University, Seoul 03760, Korea
| |
Collapse
|
24
|
Wang S, Jiang Q, Liu Y, Zhang X, Huang Y, Zhang H. The Role of Immune Cells in Moyamoya Disease. Brain Sci 2025; 15:137. [PMID: 40002470 PMCID: PMC11852451 DOI: 10.3390/brainsci15020137] [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: 12/16/2024] [Revised: 01/23/2025] [Accepted: 01/29/2025] [Indexed: 02/27/2025] Open
Abstract
Moyamoya disease (MMD) is a rare progressive cerebrovascular disorder characterized by the stenosis or occlusion of the terminal segments of the internal carotid arteries, leading to the development of abnormal collateral vascular networks. These networks are a compensatory mechanism for reduced blood flow to the brain. Despite extensive research, the exact etiology of MMD remains unknown, although recent studies suggest that immune system dysfunction plays a critical role in its pathogenesis. In particular, the involvement of immune cells such as T cells, macrophages, and dendritic cells has been increasingly recognized. These immune cells contribute to the inflammatory process and vascular remodeling observed in MMD patients, further complicating the disease's progression. Inflammation and immune-mediated damage to the vessel walls may accelerate the narrowing and occlusion of arteries, exacerbating ischemic events in the brain. Additionally, studies have revealed that certain genetic and environmental factors can influence immune system activation in MMD, linking these pathways to disease development. This review aims to provide a comprehensive overview of the immune mechanisms at play in MMD, focusing on how immune cells participate in vascular injury and remodeling. Understanding these immunological processes may offer new therapeutic targets to halt or reverse disease progression, potentially leading to more effective treatment strategies for MMD.
Collapse
Affiliation(s)
- Sheng Wang
- Department of Neurosurgery, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan 430030, China; (S.W.); (Q.J.); (Y.L.); (X.Z.); (Y.H.)
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qian Jiang
- Department of Neurosurgery, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan 430030, China; (S.W.); (Q.J.); (Y.L.); (X.Z.); (Y.H.)
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yuan Liu
- Department of Neurosurgery, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan 430030, China; (S.W.); (Q.J.); (Y.L.); (X.Z.); (Y.H.)
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xincheng Zhang
- Department of Neurosurgery, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan 430030, China; (S.W.); (Q.J.); (Y.L.); (X.Z.); (Y.H.)
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yimin Huang
- Department of Neurosurgery, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan 430030, China; (S.W.); (Q.J.); (Y.L.); (X.Z.); (Y.H.)
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Huaqiu Zhang
- Department of Neurosurgery, Tongji Hospital of Tongji Medical College of Huazhong University of Science and Technology, Jiefang Avenue 1095, Wuhan 430030, China; (S.W.); (Q.J.); (Y.L.); (X.Z.); (Y.H.)
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
25
|
Pan J, Chen S, Chen X, Song Y, Cheng H. Histone Modifications and DNA Methylation in Psoriasis: A Cellular Perspective. Clin Rev Allergy Immunol 2025; 68:6. [PMID: 39871086 DOI: 10.1007/s12016-024-09014-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/08/2024] [Indexed: 01/29/2025]
Abstract
In recent years, epigenetic modifications have attracted significant attention due to their unique regulatory mechanisms and profound biological implications. Acting as a bridge between environmental stimuli and changes in gene activity, they reshape gene expression patterns, providing organisms with regulatory mechanisms to respond to environmental changes. A growing body of evidence indicates that epigenetic regulation plays a crucial role in the pathogenesis and progression of psoriasis. A deeper understanding of these epigenetic mechanisms not only helps unveil the molecular mechanisms underlying the initiation and progression of psoriasis but may also provide new insights into diagnostic and therapeutic strategies. Given the unique roles and significant contributions of various cell types involved in the process of psoriasis, a thorough analysis of specific epigenetic patterns in different cell types becomes a key entry point for elucidating the mechanisms of disease development. Although epigenetic modifications encompass multiple complex layers, this review will focus on histone modifications and DNA methylation, describing how they function in different cell types and subsequently impact the pathophysiological processes of psoriasis. Finally, we will summarize the current problems in research concerning histone modifications and DNA methylation in psoriasis and discuss the clinical application prospects and challenges of targeting epigenetic modifications as therapeutic strategies for psoriasis.
Collapse
Affiliation(s)
- Jing Pan
- Department of Dermatology and Venereology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Siji Chen
- Department of Dermatology and Venereology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xianzhen Chen
- Department of Dermatology and Venereology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yinjing Song
- Department of Dermatology and Venereology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| | - Hao Cheng
- Department of Dermatology and Venereology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
26
|
Liu Y, Fu Q, Yang M, Xu J, Zhou Z, Chen X, Zhang Y, Yuan H, Jia G, He S, Yang L, Zhao G. Downregulation of N6-methyladenosine (m6A) methylation of Sema4D mRNA contributes to Treg dysfunction and allograft rejection. Am J Transplant 2025:S1600-6135(25)00023-1. [PMID: 39848338 DOI: 10.1016/j.ajt.2025.01.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 12/08/2024] [Accepted: 01/13/2025] [Indexed: 01/25/2025]
Abstract
Regulatory T cells (Tregs) have been shown to be involved in the induction of transplantation tolerance in numerous models. Our previous work demonstrated that methyltransferase-like 14 (METTL14) loss impaired Treg function and hindered the establishment of transplantation tolerance. However, the underlying mechanisms remain unclear. In this study, we found that METTL14 knockdown in Tregs significantly impaired their regulatory function, leading to poor allograft function and accelerated transplant rejection. Using methylated RNA immunoprecipitation- and mRNA-sequencing approaches, we discovered that METTL14 deficiency fostered the expression of semaphorin 4D (Sema4D) mRNA, a key semaphorin family member with immunoregulatory activity. Methylation of target adenosines reduced Sema4D mRNA degradation, a process mediated by the METTL14-YTH N6-methyladenosine RNA binding protein 2 axis. Inhibition of Sema4D suppressed its interaction with its receptor, thereby preserving Treg immunoregulation capability and prolonging allograft survival through the p21-activated kinase-signal transducer and activator of transcription 5signaling pathway. Importantly, Sema4D expression in kidney transplant biopsies were negatively correlated with renal allograft survival. In summary, our findings suggest that METTL14 deficiency in Tregs leads to transplant rejection and reveal for the first time that Sema4D may serve as a potential therapeutic target to enhance Treg function in transplantation.
Collapse
Affiliation(s)
- Yanzhuo Liu
- Department of Gastrointestinal Surgery, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China; Department of Oncology, Medical Research Center, Affiliated Hospital of Southwest Jiao Tong University, The Third People's Hospital of Chengdu, Chengdu, Sichuan Province, China
| | - Qiang Fu
- Organ Transplant Center, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China; Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Maozhu Yang
- Department of International Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Jianli Xu
- Institute of Neurology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Zili Zhou
- Department of Gastrointestinal Surgery, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Xingmin Chen
- Institute of Neurology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Yanling Zhang
- Department of Gastrointestinal Surgery, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China; Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Hao Yuan
- The Seventh People's Hospital of Chengdu Affiliated Cancer Hospital of Chengdu Medical College, Chengdu, Sichuan Province, China
| | - Guiqing Jia
- Department of Gastrointestinal Surgery, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Shu He
- Institute of Neurology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China
| | - Lu Yang
- Institute of Neurology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China.
| | - Gaoping Zhao
- Department of Gastrointestinal Surgery, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China; Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan Province, China.
| |
Collapse
|
27
|
Pramanik R, Chattopadhyay S, Bishayi B. Dual neutralization of TGF-β and IL-21 regulates Th17/Treg balance by suppressing inflammatory signalling in the splenic lymphocytes of Staphylococcus aureus infection-induced septic arthritic mice. Immunol Res 2025; 73:38. [PMID: 39831928 DOI: 10.1007/s12026-024-09586-2] [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: 09/25/2024] [Accepted: 12/26/2024] [Indexed: 01/22/2025]
Abstract
Septic arthritis (SA) caused by Staphylococcus aureus is a severe inflammatory joint disease, characterized by synovitis accompanied with cartilage destruction and bone erosion. The available antibiotic treatment alone is insufficient to resolve the inflammation that leads to high rates of morbidity and mortality. Among the CD4+ T helper lymphocytes, the Th17 and Tregs are key regulators of immune homeostasis. A high Th17 could lead to autoimmunity, whereas an increase in Tregs indicates immunosuppression. Depending on the external cytokine milieu, naïve CD4+ T cells transform into either Th17 or Treg cell lineage. TGF-β in the presence of IL-21 produces Th17 cells and drives the inflammatory cascade of reactions. We studied the effects of in vivo neutralization of TGF-β and IL-21 in septic arthritic mice to control arthritic inflammation, which has not been studied before. The arthritic index showed maximum severity in the SA group which substantially reduced in the Ab-treated groups. Flow cytometric analyses of peripheral blood collected from mice at 9DPI revealed the highest Th17/Treg ratio in the SA group but least in the combined-antibody-treated group. TGF-β1 and IL-21 cytokine production from serum, spleen, and synovial tissue homogenates was significantly reduced in the dual Ab-treated group than in the untreated SA group. From the Western blot analyses obtained from splenic lymphocytes at 9 DPI, we elucidated the possible underlying mechanism of interplay in downstream signalling involving the interaction between different STAT proteins and SOCS, NF-κB, RANKL, mTOR, iNOS, and COX-2 in regulating inflammation and osteoclastogenesis. On endogenous blockade with TGF-β and IL-21, the Th17/Treg ratio and resultant arthritic inflammation in SA were found to be reduced. Therefore, maintaining the Th17/Treg balance is critical to eradicate infection as well as suppress excessive inflammation and neutralization of TGF-β and IL-21 could provide a novel therapeutic strategy to treat staphylococcal SA.
Collapse
Affiliation(s)
- Rochana Pramanik
- Immunology Laboratory, Department of Physiology, University Colleges of Science and Technology, University of Calcutta, 92 APC Road, Calcutta, 700009, West Bengal, India
| | - Sreya Chattopadhyay
- Immunology Laboratory, Department of Physiology, University Colleges of Science and Technology, University of Calcutta, 92 APC Road, Calcutta, 700009, West Bengal, India
| | - Biswadev Bishayi
- Immunology Laboratory, Department of Physiology, University Colleges of Science and Technology, University of Calcutta, 92 APC Road, Calcutta, 700009, West Bengal, India.
| |
Collapse
|
28
|
Arce MM, Umhoefer JM, Arang N, Kasinathan S, Freimer JW, Steinhart Z, Shen H, Pham MTN, Ota M, Wadhera A, Dajani R, Dorovskyi D, Chen YY, Liu Q, Zhou Y, Swaney DL, Obernier K, Shy BR, Carnevale J, Satpathy AT, Krogan NJ, Pritchard JK, Marson A. Central control of dynamic gene circuits governs T cell rest and activation. Nature 2025; 637:930-939. [PMID: 39663454 PMCID: PMC11754113 DOI: 10.1038/s41586-024-08314-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 10/30/2024] [Indexed: 12/13/2024]
Abstract
The ability of cells to maintain distinct identities and respond to transient environmental signals requires tightly controlled regulation of gene networks1-3. These dynamic regulatory circuits that respond to extracellular cues in primary human cells remain poorly defined. The need for context-dependent regulation is prominent in T cells, where distinct lineages must respond to diverse signals to mount effective immune responses and maintain homeostasis4-8. Here we performed CRISPR screens in multiple primary human CD4+ T cell contexts to identify regulators that control expression of IL-2Rα, a canonical marker of T cell activation transiently expressed by pro-inflammatory effector T cells and constitutively expressed by anti-inflammatory regulatory T cells where it is required for fitness9-11. Approximately 90% of identified regulators of IL-2Rα had effects that varied across cell types and/or stimulation states, including a subset that even had opposite effects across conditions. Using single-cell transcriptomics after pooled perturbation of context-specific screen hits, we characterized specific factors as regulators of overall rest or activation and constructed state-specific regulatory networks. MED12 - a component of the Mediator complex - serves as a dynamic orchestrator of key regulators, controlling expression of distinct sets of regulators in different T cell contexts. Immunoprecipitation-mass spectrometry revealed that MED12 interacts with the histone methylating COMPASS complex. MED12 was required for histone methylation and expression of genes encoding key context-specific regulators, including the rest maintenance factor KLF2 and the versatile regulator MYC. CRISPR ablation of MED12 blunted the cell-state transitions between rest and activation and protected from activation-induced cell death. Overall, this work leverages CRISPR screens performed across conditions to define dynamic gene circuits required to establish resting and activated T cell states.
Collapse
Affiliation(s)
- Maya M Arce
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
- Biomedical Sciences graduate program, University of California, San Francisco, CA, USA
| | - Jennifer M Umhoefer
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
- Biomedical Sciences graduate program, University of California, San Francisco, CA, USA
| | - Nadia Arang
- Quantitative Biosciences Institute (QBI), University of California, San Francisco, CA, USA
| | - Sivakanthan Kasinathan
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Division of Allergy, Immunology, and Rheumatology, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Jacob W Freimer
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Zachary Steinhart
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Haolin Shen
- Biomedical Sciences graduate program, University of California, San Francisco, CA, USA
| | - Minh T N Pham
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Mineto Ota
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
| | - Anika Wadhera
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
| | - Rama Dajani
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
| | - Dmytro Dorovskyi
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Yan Yi Chen
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Qi Liu
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Yuan Zhou
- Quantitative Biosciences Institute (QBI), University of California, San Francisco, CA, USA
- Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA
| | - Danielle L Swaney
- Quantitative Biosciences Institute (QBI), University of California, San Francisco, CA, USA
- Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA
| | - Kirsten Obernier
- Quantitative Biosciences Institute (QBI), University of California, San Francisco, CA, USA
- Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA
| | - Brian R Shy
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Department of Laboratory Medicine, University of California, San Francisco, CA, USA
| | - Julia Carnevale
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, CA, USA
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Ansuman T Satpathy
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Nevan J Krogan
- Quantitative Biosciences Institute (QBI), University of California, San Francisco, CA, USA
- Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, USA
| | - Jonathan K Pritchard
- Department of Genetics, Stanford University, Stanford, CA, USA
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Alexander Marson
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA.
- Department of Medicine, University of California, San Francisco, CA, USA.
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA.
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA.
- Innovative Genomics Institute, University of California-Berkeley, Berkeley, CA, USA.
- Department of Microbiology and Immunology, University of California, San Francisco, CA, USA.
- Institute for Human Genetics, University of California, San Francisco, CA, USA.
| |
Collapse
|
29
|
Chen S, Meng G, Wang X, Jiang H, Shen M, Wang X, Wang C. Analysis of frequency changes in CD8 + regulatory T cell subsets in peripheral blood of individuals with type 1 diabetes. Diabetol Metab Syndr 2024; 16:305. [PMID: 39695694 DOI: 10.1186/s13098-024-01549-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND This study aims to examine the alterations and clinical significance of CD8+ regulatory T cell subsets in the peripheral blood of individuals with type 1 diabetes mellitus (T1DM). METHODS From January 2020 to December 2023, a study was conducted involving 40 individuals with T1DM, who visited the Department of Endocrinology at the First Affiliated Hospital of Nanjing Medical University (T1DM group). For comparison, 40 healthy individuals who underwent routine physical examinations at the same hospital during this period were selected as the control group. Peripheral blood mononuclear cells were isolated, and CD8+ T cells were labeled with CD3, CD25 and FoxP3 to analyze their subset frequencies using flow cytometry. The study examined differences in subset frequencies between the two groups and explored correlations between subset frequency, disease duration, and age of onset. RESULTS The frequencies of CD8+ CD25+, CD8+ FoxP3+, CD8+ CD25+ CD3+ and CD8+ FoxP3+ CD3+ subsets in peripheral blood mononuclear cells did not significantly differ between the healthy control group and the T1DM group (P > 0.05). In the T1DM group, the expression level of CD25 on CD8+ T cells showed no correlation with the age of onset or disease duration, and FoxP3 levels were also unrelated to the age of onset, with no statistical differences (P > 0.05). However, within the T1DM group, FoxP3 levels progressively decreased with longer disease duration, demonstrating a statistically significant negative correlation (Pearson r = -0.331, P < 0.05). In the T1DM group, the level of CD3+ CD8+ T cells expressed CD25, and there was no correlation between Foxp 3 level and age of onset, not statistically significant (P > 0.05), but the level of Foxp 3 in the T1DM group decreased with the duration of the disease, (Pearson r= - 0.363, P < 0.05). CONCLUSION The levels of CD8+FoxP3+ regulatory T cells in peripheral blood mononuclear cells of patients with T1DM show a significant correlation with disease duration, suggesting that these cells may play a critical role in the progression of T1DM.
Collapse
Affiliation(s)
- Shu Chen
- Department of Endocrinology, Nantong First People's Hospital, Affiliated Hospital 2 of Nantong University, No. 666 of Shengli Road, Chongchuan District, Nantong, 226014, Jiangsu , China
| | - Gaoqiang Meng
- Department of Neurosurgery, Nantong First People's Hospital, Affiliated Hospital 2 of Nantong University, Nantong, 226014, Jiangsu , China
| | - Xing Wang
- Department of Endocrinology, Nantong First People's Hospital, Affiliated Hospital 2 of Nantong University, No. 666 of Shengli Road, Chongchuan District, Nantong, 226014, Jiangsu , China
| | - Hemin Jiang
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu , China
| | - Min Shen
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu , China
| | - Xueqin Wang
- Department of Endocrinology, Nantong First People's Hospital, Affiliated Hospital 2 of Nantong University, No. 666 of Shengli Road, Chongchuan District, Nantong, 226014, Jiangsu , China
| | - Chunhua Wang
- Department of Endocrinology, Nantong First People's Hospital, Affiliated Hospital 2 of Nantong University, No. 666 of Shengli Road, Chongchuan District, Nantong, 226014, Jiangsu , China.
| |
Collapse
|
30
|
Li Q, Marcoux G, Hu Y, Rebetz J, Guo L, Semple E, Provan D, Xu S, Hou M, Peng J, Semple JW. Autoimmune effector mechanisms associated with a defective immunosuppressive axis in immune thrombocytopenia (ITP). Autoimmun Rev 2024; 23:103677. [PMID: 39515406 DOI: 10.1016/j.autrev.2024.103677] [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: 10/01/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
Immune thrombocytopenia (ITP) is an autoimmune disease characterized by an isolated thrombocytopenia and variable phenotype as some patients suffer no bleeding whilst others have bleeding from mild to severe, which may be fatal. This variability probably reflects the disease's complex pathophysiology; a dysregulated hyperreactive immune effector cell response involving the entire adaptive immune system (e.g. B and T cell subsets) that leads to platelet and megakaryocyte (MK) destruction. It appears that these effector responses are due to a breakdown in immune tolerance, and this is characterized by defects in several immunosuppressive cell types. These include defective T regulatory cells (Tregs), B regulatory cells (Bregs) and Myeloid-derived suppressor cells (MDSC), all of which are all intimately associated with antigen presenting cells (APC) such as dendritic cells (DC). The loss of this immunosuppressive axis allows for the activation of unchecked autoreactive T cells and B cells, leading to the development of autoantibodies and cytotoxic T cells (CTL), which can directly destroy platelets in the periphery and inhibit MK platelet production in the bone marrow (BM). This review will focus on the effector cell mechanisms in ITP and highlight the defective immunosuppressive axis that appears responsible for this platelet-specific immune hyperreactivity.
Collapse
Affiliation(s)
- Qizhao Li
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Geneviève Marcoux
- Division of Hematology and Transfusion Medicine, Lund University, Lund, Sweden
| | - Yuefen Hu
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Johan Rebetz
- Division of Hematology and Transfusion Medicine, Lund University, Lund, Sweden
| | - Li Guo
- Bloodworks Northwest Research Institute, Seattle, USA; Division of Hematology and Oncology, University of Washington; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, USA
| | | | - Drew Provan
- Department of Haematology, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, UK
| | - Shuqian Xu
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| | - Ming Hou
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Jun Peng
- Department of Hematology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - John W Semple
- Division of Hematology and Transfusion Medicine, Lund University, Lund, Sweden; Clinical Immunology and Transfusion Medicine, Office of Medical Services, Region Skåne, Lund, Sweden; Departments of Pharmacology, Medicine and Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.
| |
Collapse
|
31
|
Zou W, Sai L, Sai W, Song L, Wang G. Diagnostic and prognostic value of disulfidptosis-related genes in sepsis. INFECTIOUS MEDICINE 2024; 3:100143. [PMID: 39610785 PMCID: PMC11602581 DOI: 10.1016/j.imj.2024.100143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 07/17/2024] [Accepted: 08/26/2024] [Indexed: 11/30/2024]
Abstract
Background Sepsis is a disease associated with high morbidity and mortality rates, especially among the elderly and patients in intensive care units. Disulfidptosis, a newly identified form of cell death triggered by disulfide stress, is emerging as a significant factor in disease progression. This study aimed to explore the diagnostic and prognostic value of disulfidptosis-related genes in sepsis. Methods We obtained two datasets from the Gene Expression Omnibus (GEO) database to conduct our analysis. Functional enrichment analysis was performed to identify relevant biological pathways. A protein-protein interaction network was constructed to identify hub genes critical to sepsis. Additionally, we analyzed the immune infiltration status in sepsis patients. The diagnostic value of these hub genes for sepsis was evaluated using nomograms, receiver operating characteristic (ROC) curves, and calibration curves in both training and validation datasets. Finally, a miRNA-immune-related hub genes (miRNA-IHGs) regulatory network was developed to elucidate the synergistic interactions between miRNAs and their target genes. Results A total of 3,469 differentially expressed genes (DEGs) were identified, of which seven were related to disulfidptosis (DR-DEGs). Functional enrichment analysis showed that DR-DEGs were significantly enriched in pathways related to actin dynamics. Five hub genes (MYH10, ACTN4, MYH9, FLNA, and IQGAP1) were identified as central to these processes. The analysis of immune infiltration revealed significantly lower levels of 11 immune cell types, while macrophages and regulatory T cells were significantly elevated in sepsis patients. The area under the ROC curves (AUCs) of the IHGs risk prediction model were 0.917 and 0.894 for the training and validation sets, respectively. A miRNA-IHGs regulatory network, comprising 17 nodes and 27 edges, was constructed, with MYH9 being the most frequently regulated by miRNAs. Conclusion The pathophysiological process of sepsis appears to involve disulfidptosis, highlighting it as a potential new therapeutic targets for sepsis management.
Collapse
Affiliation(s)
- Wenlu Zou
- Department of Infectious Disease, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong Province, China
| | - Lintao Sai
- Department of Infectious Disease, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong Province, China
| | - Wen Sai
- Department of Infectious Disease, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong Province, China
| | - Li Song
- Department of Infectious Disease, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong Province, China
| | - Gang Wang
- Department of Infectious Disease, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, Shandong Province, China
| |
Collapse
|
32
|
Cauble EL, Reynolds P, Epeldegui M, Andra SS, Magpantay L, Narasimhan S, Pulivarthi D, Von Behren J, Martinez-Maza O, Goldberg D, Spielfogel ES, Lacey JV, Wang SS. Associations between per- and poly-fluoroalkyl substance (PFAS) exposure and immune responses among women in the California Teachers study: A cross-sectional evaluation. Cytokine 2024; 184:156753. [PMID: 39299102 DOI: 10.1016/j.cyto.2024.156753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 08/19/2024] [Accepted: 09/06/2024] [Indexed: 09/22/2024]
Abstract
INTRODUCTION Per- and polyfluoroalkyl substances (PFAS) are persistent environmental contaminants that have been linked to a number of health outcomes, including those related to immune dysfunction. However, there are limited numbers of epidemiological-based studies that directly examine the association between PFAS exposure and immune responses. METHODS In this cross-sectional study nested in the California Teachers Study cohort, we measured nine PFAS analytes in serum. Of the 9 analytes, we further evaluated four (PFHxS [perfluorohexane sulfonate], PFNA [perfluorononanoic acid], PFOA [perfluorooctanoic acid], PFOS [perfluorooctanesulfonic acid]) that had detection levels of > 80 %, in relation to 16 systemic inflammatory/immune markers and corresponding immune pathways (Th1 [pro-inflammatory/macrophage activation], B-cell activation, and T-cell activation). Study participants (n = 722) were female, completed a questionnaire regarding various health measures and behaviors, and donated a blood sample between 2013-2016. The association between PFAS analytes and individual immune markers and pathways were evaluated by calculating odds ratios (OR) and 95 % confidence intervals (CI) in a logistic regression model. PFAS analytes were evaluated both as a dichotomous exposure (above or below the respective median) and as a continuous variable (per 1 unit increase [ng/mL]). RESULTS The prevalence of detecting any PFAS analyte rose with increasing age, with the highest PFAS prevalence observed among those aged 75 + years and the lowest PFAS prevalence observed among those aged 40-49 years (study participant age range: 40-95 years). Significant associations with BAFF (B-cell activating factor) levels above the median were observed among participants with elevated (defined as above the median) levels of PFHxS (OR=1.53), PFOA (OR=1.43), and PFOS (OR=1.40). Similarly, there were statistically significant associations between elevated levels of PFHxS and TNFRII (tumor necrosis factor receptor 2) levels (OR=1.78) and IL2Rα (interleukin 2 receptor subunit alpha) levels (OR=1.48). We also observed significant inverse associations between elevated PFNA and sCD14 (soluble cluster of differentiation 14) (OR=0.73). No significant associations were observed between elevated PFNA and any immune marker. Evaluation of PFAS exposures as continuous exposures in association with dichotomized cytokines were generally consistent with the dichotomized associations. CONCLUSIONS PFAS exposure was associated with altered levels of circulating inflammatory/immune markers; the associations were specific to PFAS analyte and immune marker. If validated, our results may suggest potential immune mechanisms underlying associations between the different PFAS analytes and adverse health outcomes.
Collapse
Affiliation(s)
- Emily L Cauble
- Division of Health Analytics, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Peggy Reynolds
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Marta Epeldegui
- UCLA AIDS Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Syam S Andra
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Srinivasan Narasimhan
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Divya Pulivarthi
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Julie Von Behren
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | | | - Debbie Goldberg
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
| | - Emma S Spielfogel
- Division of Health Analytics, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - James V Lacey
- Division of Health Analytics, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Sophia S Wang
- Division of Health Analytics, Beckman Research Institute, City of Hope, Duarte, CA, USA.
| |
Collapse
|
33
|
Zhao Y, Tan M, Yin Y, Zhang J, Song Y, Li H, Yan L, Jin Y, Wu Z, Yang T, Jiang T, Li H. Comprehensive macro and micro views on immune cells in ischemic heart disease. Cell Prolif 2024; 57:e13725. [PMID: 39087342 PMCID: PMC11628753 DOI: 10.1111/cpr.13725] [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: 01/07/2024] [Revised: 06/25/2024] [Accepted: 07/18/2024] [Indexed: 08/02/2024] Open
Abstract
Ischemic heart disease (IHD) is a prevalent cardiovascular condition that remains the primary cause of death due to its adverse ventricular remodelling and pathological changes in end-stage heart failure. As a complex pathologic condition, it involves intricate regulatory processes at the cellular and molecular levels. The immune system and cardiovascular system are closely interconnected, with immune cells playing a crucial role in maintaining cardiac health and influencing disease progression. Consequently, alterations in the cardiac microenvironment are influenced and controlled by various immune cells, such as macrophages, neutrophils, dendritic cells, eosinophils, and T-lymphocytes, along with the cytokines they produce. Furthermore, studies have revealed that Gata6+ pericardial cavity macrophages play a key role in regulating immune cell migration and subsequent myocardial tissue repair post IHD onset. This review outlines the role of immune cells in orchestrating inflammatory responses and facilitating myocardial repair following IHD, considering both macro and micro views. It also discusses innovative immune cell-based therapeutic strategies, offering new insights for further research on the pathophysiology of ischemic heart disease and immune cell-targeted therapy for IHD.
Collapse
Affiliation(s)
- Yongjian Zhao
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Mingyue Tan
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
- Department of Geriatrics, Southwest HospitalThe Third Military Medical University (Army Medical University)ChongqingChina
| | - Yunfei Yin
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Jun Zhang
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Yiyi Song
- Suzhou Medical College of Soochow UniversityJiangsuChina
| | - Hang Li
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Lin Yan
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Yifeng Jin
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Ziyue Wu
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Tianke Yang
- Department of Ophthalmology, The First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefeiChina
| | - Tingbo Jiang
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| | - Hongxia Li
- Department of CardiologyThe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuChina
| |
Collapse
|
34
|
Gao C, Chen L, Xie XY, He XF, Shen J, Zheng L. Bone marrow mesenchymal stem cells-derived exosomal miR-381 alleviates lung ischemia-reperfusion injury by activating Treg differentiation through inhibiting YTHDF1 expression. Cell Signal 2024; 124:111440. [PMID: 39357613 DOI: 10.1016/j.cellsig.2024.111440] [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: 06/17/2024] [Revised: 09/09/2024] [Accepted: 09/29/2024] [Indexed: 10/04/2024]
Abstract
AIM Our study aimed to investigate whether BMSCs-derived exosomal miR-381 promotes Treg cell differentiation in lung ischemia-reperfusion injury (LIRI), and the underlying mechanism. METHODS The in vitro and in vivo models of LIRI were established by hypoxia/reoxygenation (H/R) treatment and lung ischemia/reperfusion (I/R) surgery, respectively. BMSCs-derived exosomes were isolated and identified by western blot, nanoparticle tracking analysis, and transmission electron microscopy. Cell viability, proliferation, and apoptosis were assessed by CCK-8, EdU, and flow cytometry assay, respectively. IL-18 secretion level in lung microvascular endothelial cells (LMECs) and lung tissue homogenate was examined by ELISA. Treg cell differentiation was determined using flow cytometry. The relationships between miR-381, YTHDF1, and IL-18 were investigated using dual-luciferase reporter gene, RIP, and/or RNA pull-down assays. MeRIP assay was employed to determine m6A modification of IL-18 mRNA in LMECs. The ubiquitination level of Foxp3 protein in CD4+ T cells was analyzed by Co-IP assay. RESULTS BMSCs-derived exosomes reduced LMECs injury and increased Treg cell differentiation in LIRI, whereas miR-381 inhibition in BMSCs weakened these impacts. Mechanistically, miR-381 inhibited IL-18 translation in LMECs by inhibiting YTHDF1 expression via binding to its 3'-UTR. As expected, YTHDF1 overexpression in LMECs abolished the effects of miR-381-overexpressed exosomes on LMECs injury and Treg cell differentiation. Moreover, LMECs-secreted IL-18 inhibited Treg cell differentiation by promoting the ubiquitination degradation of Foxp3 protein. CONCLUSION BMSCs-derived exosomal miR-381 suppressed IL-18 translation in LMECs through binding to YTHDF1 3'-UTR, thus suppressing the ubiquitination degradation of Foxp3 in CD4+ T cells, which promoted Treg cell differentiation and mitigated LIRI development.
Collapse
Affiliation(s)
- Cao Gao
- Departments of Anesthesiology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, Jiangsu, China
| | - Lei Chen
- Department of Thoracic Surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213003, Jiangsu, China
| | - Xiang-Yu Xie
- Department of Thoracic Surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213003, Jiangsu, China
| | - Xiao-Feng He
- Departments of Anesthesiology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, Jiangsu, China
| | - Jiang Shen
- Departments of Anesthesiology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, Jiangsu, China
| | - Liang Zheng
- Department of Thoracic Surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213003, Jiangsu, China.
| |
Collapse
|
35
|
Liu Z, Ke SR, Shi ZX, Zhou M, Sun L, Sun QH, Xiao B, Wang DL, Huang YJ, Lin JS, Wang HS, Zhang QK, Pan CN, Liang XW, Chen RX, Mao Z, Lin XC. Dynamic transition of Tregs to cytotoxic phenotype amid systemic inflammation in Graves' ophthalmopathy. JCI Insight 2024; 9:e181488. [PMID: 39365735 PMCID: PMC11601897 DOI: 10.1172/jci.insight.181488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 09/26/2024] [Indexed: 10/06/2024] Open
Abstract
Graves' disease (GD) is an autoimmune condition that can progress to Graves' ophthalmopathy (GO), leading to irreversible damage to orbital tissues and potential blindness. The pathogenic mechanism is not fully understood. In this study, we conducted single-cell multi-omics analyses on healthy individuals, patients with GD without GO, newly diagnosed patients with GO, and treated patients with GO. Our findings revealed gradual systemic inflammation during GO progression, marked by overactivation of cytotoxic effector T cell subsets, and expansion of specific T cell receptor clones. Importantly, we observed a decline in the immunosuppressive function of activated Treg (aTreg) accompanied by a cytotoxic phenotypic transition. In vitro experiments revealed that dysfunction and transition of GO-autoreactive Treg were regulated by the yin yang 1 (YY1) upon secondary stimulation of thyroid stimulating hormone receptor (TSHR) under inflammatory conditions. Furthermore, adoptive transfer experiments of the GO mouse model confirmed infiltration of these cytotoxic Treg into the orbital lesion tissues. Notably, these cells were found to upregulate inflammation and promote pathogenic fibrosis of orbital fibroblasts (OFs). Our results reveal the dynamic changes in immune landscape during GO progression and provide direct insights into the instability and phenotypic transition of Treg, offering potential targets for therapeutic intervention and prevention of autoimmune diseases.
Collapse
|
36
|
Hu W, Zhang X, Wu Z, Luo Y, Hu B, Zou X. Exploring and Validating the Mechanism of Ulinastatin in the Treatment of Sepsis-Associated Encephalopathy Based on Transcriptome Sequencing. J Inflamm Res 2024; 17:8753-8773. [PMID: 39564549 PMCID: PMC11573691 DOI: 10.2147/jir.s488400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 11/07/2024] [Indexed: 11/21/2024] Open
Abstract
Purpose Sepsis can induce sepsis-associated encephalopathy (SAE), with Ulinastatin (UTI) serving a critical anti-inflammatory role. This study aimed to identify the hub genes in an SAE mouse model following UTI intervention and investigate the underlying molecular mechanisms. Materials and Methods Through differential expression analysis to obtain differentially expressed genes (DEGs), ie, UTI vs CLP (DEGs1) and Con vs CLP (DEGs2). After taking the intersection of the genes with opposite differential trends in these two parts and immune-related genes (IRGs), DE-IRGs were obtained. Hub genes in the protein-protein interaction (PPI) network were then determined using six algorithms from the Cytohubba plugin in Cytoscape. Gene set enrichment analysis (GSEA) was employed to explore the functional relevance of these hub genes. Additionally, the immune microenvironment across the three groups was compared, and hub gene-related drugs were predicted using an online database. Finally, qRT-PCR was used to validate the expression of the hub genes in hippocampal tissue from CLP mice. Results RNA sequencing obtained 864 differentially expressed genes (DEGs) (CLP vs Con) and 279 DEGs (UTI vs CLP). Taking the intersection of DEGs with opposite expression trends yielded 165 DEGs. Six key genes (ICAM - 1, IRF7, IL - 1β, CCL2, IL - 6 and SOCS3) were screened by six algorithms. Immune infiltration analysis found that Treg cells were reversed after treatment with UTI in the diseased state. A total of 106 hub - gene - related drugs were predicted, among which BINDARIT - CCL2 and LIFITEGRAST - ICAM1 showed particularly high affinities. The qRT - PCR verification results were consistent with the sequencing results. Conclusion In conclusion, ICAM-1, IRF7, IL-1β, CCL2, IL-6, and SOCS3 were identified as potential therapeutic targets in SAE mice treated with UTI. This study offers theoretical support for UTI as a treatment option for SAE.
Collapse
Affiliation(s)
- Wen Hu
- Guizhou Medical University, Guiyang, Guizhou, 550004, People's Republic of China
| | - Xiaoyuan Zhang
- Guizhou Medical University, Guiyang, Guizhou, 550004, People's Republic of China
| | - Zhen Wu
- Guizhou Medical University, Guiyang, Guizhou, 550004, People's Republic of China
| | - Yushan Luo
- Guizhou Medical University, Guiyang, Guizhou, 550004, People's Republic of China
| | - Bailong Hu
- Department of Anesthesiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, People's Republic of China
| | - Xiaohua Zou
- Department of Anesthesiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, People's Republic of China
| |
Collapse
|
37
|
Mu S, Wang W, Liu Q, Ke N, Li H, Sun F, Zhang J, Zhu Z. Autoimmune disease: a view of epigenetics and therapeutic targeting. Front Immunol 2024; 15:1482728. [PMID: 39606248 PMCID: PMC11599216 DOI: 10.3389/fimmu.2024.1482728] [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: 08/23/2024] [Accepted: 10/23/2024] [Indexed: 11/29/2024] Open
Abstract
Autoimmune diseases comprise a large group of conditions characterized by a complex pathogenesis and significant heterogeneity in their clinical manifestations. Advances in sequencing technology have revealed that in addition to genetic susceptibility, various epigenetic mechanisms including DNA methylation and histone modification play critical roles in disease development. The emerging field of epigenetics has provided new perspectives on the pathogenesis and development of autoimmune diseases. Aberrant epigenetic modifications can be used as biomarkers for disease diagnosis and prognosis. Exploration of human epigenetic profiles revealed that patients with autoimmune diseases exhibit markedly altered DNA methylation profiles compared with healthy individuals. Targeted cutting-edge epigenetic therapies are emerging. For example, DNA methylation inhibitors can rectify methylation dysregulation and relieve patients. Histone deacetylase inhibitors such as vorinostat can affect chromatin accessibility and further regulate gene expression, and have been used in treating hematological malignancies. Epigenetic therapies have opened new avenues for the precise treatment of autoimmune diseases and offer new opportunities for improved therapeutic outcomes. Our review can aid in comprehensively elucidation of the mechanisms of autoimmune diseases and development of new targeted therapies that ultimately benefit patients with these conditions.
Collapse
Affiliation(s)
- Siqi Mu
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China
- Department of Skin Genetics, Anhui Province Laboratory of Inflammation and Immune Mediated Diseases, Hefei, Anhui, China
- Department of Dermatology, Shannan People's Hospital, Shannan, China
- First Clinical Medical College, Anhui Medical University, Hefei, Anhui, China
| | - Wanrong Wang
- Department of Skin Genetics, Anhui Province Laboratory of Inflammation and Immune Mediated Diseases, Hefei, Anhui, China
- Department of Dermatology, Shannan People's Hospital, Shannan, China
- First Clinical Medical College, Anhui Medical University, Hefei, Anhui, China
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Pharmacology, Basic Medical College, Anhui Medical University, Hefei, Anhui, China
| | - Qiuyu Liu
- First Clinical Medical College, Anhui Medical University, Hefei, Anhui, China
| | - Naiyu Ke
- Department of Ophthalmology, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Hao Li
- Department of Urology, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Feiyang Sun
- First Clinical Medical College, Anhui Medical University, Hefei, Anhui, China
| | - Jiali Zhang
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China
- Department of Skin Genetics, Anhui Province Laboratory of Inflammation and Immune Mediated Diseases, Hefei, Anhui, China
- Department of Dermatology, Shannan People's Hospital, Shannan, China
| | - Zhengwei Zhu
- Department of Dermatology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, Anhui, China
- Department of Skin Genetics, Anhui Province Laboratory of Inflammation and Immune Mediated Diseases, Hefei, Anhui, China
- Department of Dermatology, Shannan People's Hospital, Shannan, China
| |
Collapse
|
38
|
Singer M, Elsayed AM, Husseiny MI. Regulatory T-cells: The Face-off of the Immune Balance. FRONT BIOSCI-LANDMRK 2024; 29:377. [PMID: 39614434 DOI: 10.31083/j.fbl2911377] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/29/2024] [Accepted: 08/13/2024] [Indexed: 12/01/2024]
Abstract
Regulatory T-cells (Tregs) play a crucial role in maintaining immune homeostasis, ensuring a balanced immune response. Tregs primarily operate in an antigen-specific fashion, facilitated by their distinct distribution within discrete niches. Tregs have been studied extensively, from their point of origin in the thymus origin to their fate in the periphery or organs. Signals received from antigen-presenting cells (APCs) stimulate Tregs to dampen inflammation. Almost all tumors are characterized by a pathological abundance of immune suppression in their microenvironment. Conversely, the lack thereof proves detrimental to immunological disorders. Achieving a balanced expression of Tregs in relation to other immune compartments is important in establishing an effective and adaptable immune tolerance towards cancer cells and autoantigens. In the context of cancer, it is essential to decrease the frequency of Tregs to overcome tumor suppression. A lower survival rate is associated with the presence of excessive exhausted effector immune cells and an increased frequency of regulatory cells. However, when it comes to treating graft rejection and autoimmune diseases, the focus lies on immune tolerance and the transfer of Tregs. Here, we explore the complex mechanisms that Tregs use in human disease to balance effector immune cells.
Collapse
Affiliation(s)
- Mahmoud Singer
- School of Medicine, University of California Irvine, Irvine, CA 92617, USA
| | - Ahmed M Elsayed
- Division of Infectious Diseases, The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Mohamed I Husseiny
- Department of Translational Research & Cellular Therapeutics, Arthur Riggs Diabetes & Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
39
|
Zhang A, Fan T, Liu Y, Yu G, Li C, Jiang Z. Regulatory T cells in immune checkpoint blockade antitumor therapy. Mol Cancer 2024; 23:251. [PMID: 39516941 PMCID: PMC11545879 DOI: 10.1186/s12943-024-02156-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024] Open
Abstract
Regulatory T cells (Tregs), an essential component of the human immune system, are a heterogeneous group of T lymphocytes with the ability to suppress immune responses and maintain immune homeostasis. Recent evidence indicates that Tregs may impair antitumor immunity and facilitate cancer progression by weakening functions of effector T cells (Teffs). Consequently, targeting Tregs to eliminate them from tumor microenvironments to improve Teffs' activity could emerge as an effective strategy for cancer immunotherapy. This review outlines the biology of Tregs, detailing their origins, classification, and crucial markers. Our focus lies on the complex role of Tregs in cancer's development, progression and treatment, particularly on their suppressive role upon antitumor responses via multiple mechanisms. We delve into Tregs' involvement in immune checkpoint blockade (ICB) therapy, their dual effect on cancer immunotherapy and their potential biomarkers for ICB therapy effectiveness. We also summarize advances in the therapies that adjust Tregs to optimize ICB therapy, which may be crucial for devising innovative cancer treatment strategies.
Collapse
Affiliation(s)
- An Zhang
- Department of Colorectal Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Tao Fan
- Department of Thoracic Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yixiao Liu
- Department of Colorectal Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Guanhua Yu
- Department of Colorectal Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chunxiang Li
- Department of Thoracic Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Zheng Jiang
- Department of Colorectal Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
40
|
Raposo AASF, Rosmaninho P, Silva SL, Paço S, Brazão ME, Godinho-Santos A, Tokunaga-Mizoro Y, Nunes-Cabaço H, Serra-Caetano A, Almeida ARM, Sousa AE. Decoding mutational hotspots in human disease through the gene modules governing thymic regulatory T cells. Front Immunol 2024; 15:1458581. [PMID: 39483472 PMCID: PMC11525063 DOI: 10.3389/fimmu.2024.1458581] [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: 07/02/2024] [Accepted: 09/17/2024] [Indexed: 11/03/2024] Open
Abstract
Computational strategies to extract meaningful biological information from multiomics data are in great demand for effective clinical use, particularly in complex immune-mediated disorders. Regulatory T cells (Tregs) are essential for immune homeostasis and self-tolerance, controlling inflammatory and autoimmune processes in many diseases with a multigenic basis. Here, we quantify the Transcription Factor (TF) differential occupancy landscape to uncover the Gene Regulatory Modules governing lineage-committed Tregs in the human thymus, and show that it can be used as a tool to prioritise variants in complex diseases. We combined RNA-seq and ATAC-seq and generated a matrix of differential TF binding to genes differentially expressed in Tregs, in contrast to their counterpart conventional CD4 single-positive thymocytes. The gene loci of both established and novel genetic interactions uncovered by the Gene Regulatory Modules were significantly enriched in rare variants carried by patients with common variable immunodeficiency, here used as a model of polygenic-based disease with severe inflammatory and autoimmune manifestations. The Gene Regulatory Modules controlling the Treg signature can, therefore, be a valuable resource for variant classification, and to uncover new therapeutic targets. Overall, our strategy can also be applied in other biological processes of interest to decipher mutational hotspots in individual genomes.
Collapse
Affiliation(s)
- Alexandre A. S. F. Raposo
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Pedro Rosmaninho
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Susana L. Silva
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Serviço de Imunoalergologia, Hospital de Santa Maria, Unidade Local de Saúde (ULS) Santa Maria, Lisboa, Portugal
| | - Susana Paço
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Maria E. Brazão
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Ana Godinho-Santos
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Yumie Tokunaga-Mizoro
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Helena Nunes-Cabaço
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Ana Serra-Caetano
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Afonso R. M. Almeida
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Ana E. Sousa
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
41
|
Iesari S, Nava FL, Zais IE, Coubeau L, Ferraresso M, Favi E, Lerut J. Advancing immunosuppression in liver transplantation: A narrative review. Hepatobiliary Pancreat Dis Int 2024; 23:441-448. [PMID: 38523030 DOI: 10.1016/j.hbpd.2024.03.001] [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: 05/08/2023] [Accepted: 03/14/2024] [Indexed: 03/26/2024]
Abstract
Immunosuppression is essential to ensure recipient and graft survivals after liver transplantation (LT). However, our understanding and management of the immune system remain suboptimal. Current immunosuppressive therapy cannot selectively inhibit the graft-specific immune response and entails a significant risk of serious side effects, i.e., among others, de novo cancers, infections, cardiovascular events, renal failure, metabolic syndrome, and late graft fibrosis, with progressive loss of graft function. Pharmacological research, aimed to develop alternative immunosuppressive agents in LT, is behind other solid-organ transplantation subspecialties, and, therefore, the development of new compounds and strategies should get priority in LT. The research trajectories cover mechanisms to induce T-cell exhaustion, to inhibit co-stimulation, to mitigate non-antigen-specific inflammatory response, and, lastly, to minimize the development and action of donor-specific antibodies. Moreover, while cellular modulation techniques are complex, active research is underway to foster the action of T-regulatory cells, to induce tolerogenic dendritic cells, and to promote the function of B-regulatory cells. We herein discuss current lines of research in clinical immunosuppression, particularly focusing on possible applications in the LT setting.
Collapse
Affiliation(s)
- Samuele Iesari
- General Surgery and Kidney Transplantation, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 15 Via della Commenda, 20122 Milan, Italy
| | - Francesca Laura Nava
- General Surgery and Kidney Transplantation, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 15 Via della Commenda, 20122 Milan, Italy
| | - Ilaria Elena Zais
- General Surgery and Kidney Transplantation, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 15 Via della Commenda, 20122 Milan, Italy
| | - Laurent Coubeau
- Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, 10 Avenue Hippocrate, 1200 Brussels, Belgium; Service de Chirurgie et Transplantation Abdominale, Cliniques Universitaires Saint-Luc, 55 Avenue Hippocrate, 1200 Brussels, Belgium
| | - Mariano Ferraresso
- General Surgery and Kidney Transplantation, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 15 Via della Commenda, 20122 Milan, Italy; Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 19 Via della Commenda, 20122 Milan, Italy
| | - Evaldo Favi
- General Surgery and Kidney Transplantation, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 15 Via della Commenda, 20122 Milan, Italy; Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 19 Via della Commenda, 20122 Milan, Italy.
| | - Jan Lerut
- Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, 10 Avenue Hippocrate, 1200 Brussels, Belgium
| |
Collapse
|
42
|
Xie WY, Shen HL, Yan ZM, Zheng RJ, Jiang JJ, Zhong JJ, Zhou WW. Paenibacillus exopolysaccharide alleviates Malassezia-induced skin damage: Enhancing skin barrier function, regulating immune responses, and modulating microbiota. Int J Biol Macromol 2024; 278:135404. [PMID: 39256124 DOI: 10.1016/j.ijbiomac.2024.135404] [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: 12/29/2023] [Revised: 08/09/2024] [Accepted: 09/05/2024] [Indexed: 09/12/2024]
Abstract
Numerous studies have established a strong association between Malassezia and various skin disorders, including atopic dermatitis. Finding appropriate methods or medications to alleviate Malassezia-induced skin damage is of notable public interest. This study aimed to evaluate the therapeutic effect of the exopolysaccharide EPS1, produced by Paenibacillus polymyxa, on Malassezia restricta-induced skin damage. In vitro assays indicated that EPS1 reduced the expression of pro-inflammatory cytokine genes in TNF-α-induced HaCaT cells. In a murine model, EPS1 was found to mitigate clinical symptoms, reduce epidermal thickness and mast cell infiltration, improve skin barrier function, decrease pro-inflammatory cytokine levels associated with type 17 inflammation, enhance Tregs in the spleen, upregulate the transcription of Treg-related genes in skin lesions, and modulate the skin microbiota. This study is the first to report the alleviating effect of Paenibacillus exopolysaccharide on Malassezia-induced skin inflammation and its impact on the skin microbiota. These findings support the potential of Paenibacillus exopolysaccharides as consumer products and therapeutic agents for managing Malassezia-induced skin damage by improving skin barrier function, modulating immune responses, and influencing skin microbiota.
Collapse
Affiliation(s)
- Wan-Yue Xie
- Institute of Food Bioscience and Technology, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Hui-Ling Shen
- Institute of Food Bioscience and Technology, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Zi-Ming Yan
- Institute of Food Bioscience and Technology, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, Zhejiang, China
| | - Ru-Jing Zheng
- Zhejiang Homay Technology Co., Ltd., Hangzhou 311200, Zhejiang, China
| | - Jin-Jie Jiang
- Zhejiang Homay Technology Co., Ltd., Hangzhou 311200, Zhejiang, China
| | - Jian-Jiang Zhong
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Wen-Wen Zhou
- Institute of Food Bioscience and Technology, College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, Zhejiang, China.
| |
Collapse
|
43
|
Xue D, Guo X, Liu J, Li Y, Liu L, Liao G, Zhang M, Cao J, Liu Y, Lou J, Li H, Mi W, Wang L, Fu Q. Tryptophan-rich diet and its effects on Htr7 + Tregs in alleviating neuroinflammation and cognitive impairment induced by lipopolysaccharide. J Neuroinflammation 2024; 21:241. [PMID: 39334486 PMCID: PMC11437714 DOI: 10.1186/s12974-024-03239-9] [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: 01/23/2024] [Accepted: 09/23/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Neuroinflammation is a vital pathogenic mechanism for neurodegenerative diseases such as Alzheimer's, schizophrenia, and age-related cognitive decline. Regulatory T cells (Tregs) exhibit potent anti-inflammatory properties and can modulate neurodegenerative diseases arising from central nervous system inflammatory responses. However, the role of Tregs in neuroinflammation-related cognitive dysfunction remains unclear. It is highly plausible that Htr7+ Tregs expressing unique genes associated with the nervous system, including the Htr7 gene encoding the serotonin receptor 5-HT7, play a pivotal role. METHODS Mice were given a tryptophan-rich diet (with a tryptophan content of 0.6%) or a normal diet (with a tryptophan content of 0.16%). The neuroinflammation-mediated cognitive dysfunction model was established by intracerebroventricular injection of lipopolysaccharide (LPS) in 8-week-old C57BL/6J mice. The activation and infiltration of Tregs were measured using flow cytometry. Primary Tregs were cocultured separately with primary CD8+ T cells and primary microglia for in vitro validation of the impact of 5-HT and 5-HT7 receptor on Tregs. Prior to their transfer into recombination activating gene 1 (Rag1-/-) mice, Tregs were ex vivo transfected with lentivirus to knock down the expression of Htr7. RESULTS In this study, the tryptophan-rich diet was found to reverse LPS-induced cognitive impairment and reduce the levels of 5-HT in peripheral blood. The tryptophan-rich diet led to increased levels of 5-HT in peripheral blood, which in turn promoted the proliferation and activation of Htr7+ Tregs. Additionally, the tryptophan-rich diet was also shown to attenuate LPS-mediated neuroinflammation by activating Htr7+ Tregs. Furthermore, 5-HT and 5-HT7 receptor were found to enhance the immunosuppressive effect of Tregs on CD8+ T cells and microglia. In Rag1-/- mice, Htr7+ Tregs were shown to alleviate LPS-induced neuroinflammation and cognitive impairment. CONCLUSIONS Our research revealed the ability of Htr7+ Tregs to mitigate neuroinflammation and prevent neuronal damage by suppressing the infiltration of CD8+ T cells into the brain and excessive activation of microglia, thereby ameliorating LPS-induced cognitive impairment. These insights may offer novel therapeutic targets involving Tregs for neuroinflammation and cognitive impairment.
Collapse
Affiliation(s)
- Dinghao Xue
- Department of Anesthesiology, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
- Department of Anesthesiology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Xu Guo
- Department of Anesthesiology, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Jingjing Liu
- Department of Anesthesiology, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
- Department of Anesthesiology, Chinese People's Armed Police Force Hospital of Beijing, Beijing, 100027, China
| | - Yanxiang Li
- Department of Anesthesiology, The 71st Group Army Hospital of CPLA Army, Xuzhou, 221004, China
| | - Luyu Liu
- Department of Anesthesiology, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Guosong Liao
- Department of Anesthesiology, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Mingru Zhang
- Department of Anesthesiology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Jiangbei Cao
- Department of Anesthesiology, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yanhong Liu
- Department of Anesthesiology, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Jingsheng Lou
- Department of Anesthesiology, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Hao Li
- Department of Anesthesiology, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Weidong Mi
- Department of Anesthesiology, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Long Wang
- Department of Pain Medicine, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Qiang Fu
- Department of Anesthesiology, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
44
|
Golden GJ, Wu VH, Hamilton JT, Amses KR, Shapiro MR, Japp AS, Liu C, Pampena MB, Kuri-Cervantes L, Knox JJ, Gardner JS, Atkinson MA, Brusko TM, Prak ETL, Kaestner KH, Naji A, Betts MR. Immune perturbations in human pancreas lymphatic tissues prior to and after type 1 diabetes onset. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.23.590798. [PMID: 39345402 PMCID: PMC11429609 DOI: 10.1101/2024.04.23.590798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Autoimmune destruction of pancreatic β cells results in type 1 diabetes (T1D), with pancreatic immune infiltrate representing a key feature in this process. Studies of human T1D immunobiology have predominantly focused on circulating immune cells in the blood, while mouse models suggest diabetogenic lymphocytes primarily reside in pancreas-draining lymph nodes (pLN). A comprehensive study of immune cells in human T1D was conducted using pancreas draining lymphatic tissues, including pLN and mesenteric lymph nodes, and the spleen from non-diabetic control, β cell autoantibody positive non-diabetic (AAb+), and T1D organ donors using complementary approaches of high parameter flow cytometry and CITEseq. Immune perturbations suggestive of a proinflammatory environment were specific for T1D pLN and AAb+ pLN. In addition, certain immune populations correlated with high T1D genetic risk independent of disease state. These datasets form an extensive resource for profiling human lymphatic tissue immune cells in the context of autoimmunity and T1D.
Collapse
Affiliation(s)
- Gregory J Golden
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Vincent H Wu
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Jacob T Hamilton
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Kevin R Amses
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Melanie R Shapiro
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, College of Medicine, Gainesville, FL 32610, USA
| | - Alberto Sada Japp
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Chengyang Liu
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Department of Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Maria Betina Pampena
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Leticia Kuri-Cervantes
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - James J Knox
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Jay S Gardner
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Mark A Atkinson
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, College of Medicine, Gainesville, FL 32610, USA
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Todd M Brusko
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, College of Medicine, Gainesville, FL 32610, USA
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Eline T Luning Prak
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Klaus H Kaestner
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Ali Naji
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Department of Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Michael R Betts
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| |
Collapse
|
45
|
Yu X, Li S, Zhong J, Ji X, Xu H, Chen Q, Li Q. Single-cell transcriptome sequencing partially revealed the changes of T cells in the early stage of aging kidney. Mol Immunol 2024; 173:61-70. [PMID: 39059207 DOI: 10.1016/j.molimm.2024.06.005] [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: 01/09/2024] [Revised: 04/23/2024] [Accepted: 06/14/2024] [Indexed: 07/28/2024]
Abstract
Aging is a gradual, inevitable physiologic process. The organ aging is related to the persistence of chronic inflammation, but the understanding of inflammatory state during renal aging is lacking currently. Single-cell transcriptome sequencing was performed on aging mouse kidney to reveal the molecular phenotype and composition changes of different cell types. In the early stage of aging, immune cells such as T, B cells and mononuclear macrophages increased in kidney. The molecular state of T cells in aging kidney changed and polarized. Among them, we identified a group of GZMK+ CD8 + T cells with high expression of Eomes, Pdcd1 and Ifng and a group of Il17a+ T cells with high expression of Il17a and Il23r. Moreover, the cytokines and inflammations can aggravate tissue damage eventually. Furthermore, we found the interaction between different types of epithelial cells and T cells increased during the renal aging. These results identify the changes of T cells in the early stage of aging kidney and suggest that GZMK+CD8+ T cells might be a potential target to ameliorate age-associated dysfunctions of kidney(Graphical Abstract).
Collapse
Affiliation(s)
- Xinyi Yu
- Department of Nephrology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
| | - Shuying Li
- Department of Nephrology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
| | - Jinjie Zhong
- Department of Nephrology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China
| | - Xiaoqian Ji
- Institute for Advanced Study and School of Physical Science and Technology, Soochow University, Suzhou, Jiangsu, China
| | - Huizhong Xu
- Institute for Advanced Study and School of Physical Science and Technology, Soochow University, Suzhou, Jiangsu, China
| | - Qilin Chen
- Department of Nephrology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.
| | - Qiu Li
- Department of Nephrology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing, China.
| |
Collapse
|
46
|
Lang HP, Osum KC, Friedenberg SG. A review of CD4 + T cell differentiation and diversity in dogs. Vet Immunol Immunopathol 2024; 275:110816. [PMID: 39173398 PMCID: PMC11421293 DOI: 10.1016/j.vetimm.2024.110816] [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: 06/19/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 08/24/2024]
Abstract
CD4+ T cells are an integral component of the adaptive immune response, carrying out many functions to combat a diverse range of pathogenic challenges. These cells exhibit remarkable plasticity, differentiating into specialized subsets such as T helper type 1 (TH1), TH2, TH9, TH17, TH22, regulatory T cells (Tregs), and follicular T helper (TFH) cells. Each subset is capable of addressing a distinct immunological need ranging from pathogen eradication to regulation of immune homeostasis. As the immune response subsides, CD4+ T cells rest down into long-lived memory phenotypes-including central memory (TCM), effector memory (TEM), resident memory (TRM), and terminally differentiated effector memory cells (TEMRA) that are localized to facilitate a swift and potent response upon antigen re-encounter. This capacity for long-term immunological memory and rapid reactivation upon secondary exposure highlights the role CD4+ T cells play in sustaining both adaptive defense mechanisms and maintenance. Decades of mouse, human, and to a lesser extent, pig T cell research has provided the framework for understanding the role of CD4+ T cells in immune responses, but these model systems do not always mimic each other. Although our understanding of pig immunology is not as extensive as mouse or human research, we have gained valuable insight by studying this model. More akin to pigs, our understanding of CD4+ T cells in dogs is much less complete. This disparity exists in part because canine immunologists depend on paradigms from mouse and human studies to characterize CD4+ T cells in dogs, with a fraction of available lineage-defining antibody markers. Despite this, every major CD4+ T cell subset has been described to some extent in dogs. These subsets have been studied in various contexts, including in vitro stimulation, homeostatic conditions, and across a range of disease states. Canine CD4+ T cells have been categorized according to lineage-defining characteristics, trafficking patterns, and what cytokines they produce upon stimulation. This review addresses our current understanding of canine CD4+ T cells from a comparative perspective by highlighting both the similarities and differences from mouse, human, and pig CD4+ T cell biology. We also discuss knowledge gaps in our current understanding of CD4+ T cells in dogs that could provide direction for future studies in the field.
Collapse
Affiliation(s)
- Haeree P Lang
- Center for Immunology, University of Minnesota, Minneapolis, MN 55414, USA; Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA.
| | - Kevin C Osum
- Center for Immunology, University of Minnesota, Minneapolis, MN 55414, USA.
| | - Steven G Friedenberg
- Center for Immunology, University of Minnesota, Minneapolis, MN 55414, USA; Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN 55108, USA.
| |
Collapse
|
47
|
Li T, Wu T, Li X, Qian C. Transcriptional switches in melanoma T Cells: Facilitating polarizing into regulatory T cells. Int Immunopharmacol 2024; 137:112484. [PMID: 38885605 DOI: 10.1016/j.intimp.2024.112484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 06/11/2024] [Accepted: 06/11/2024] [Indexed: 06/20/2024]
Abstract
Melanoma is a malignant skin tumor with a high mortality rate. Regulatory T cells (Tregs) are immune cells with immunosuppressive roles, however, the precise mechanisms governing Treg involvement in melanoma remain enigmatic. Experimental findings unveiled different transcription factor switches between normal and tumor T cell, with heightened FOXP3 and BATF in the latter. These factors induced immunosuppressive molecules and Treg maintenance genes, polarizing tumor T cells into Tregs. Spatial transcriptomics illuminated the preferential settlement of Tregs at the melanoma periphery. Within this context, FOXP3 in Tregs facilitated direct enhancement of specific ligand gene expression, fostering communication with neighboring cells. Novel functional molecules bound to FOXP3 or BATF in Tregs, such as SPOCK2, SH2D2A, and ligand molecules ITGB2, LTA, CLEC2C, CLEC2D, were discovered, which had not been previously reported in melanoma Treg studies. Furthermore, we validated our findings in a large number of clinical samples and identified the Melanoma Treg-Specific Regulatory Tag Set (Mel TregS). ELISA analysis showed that the protein levels of Mel TregS in melanoma Tregs were higher than in normal Tregs. We then utilized SERS technology to measure the signal values of Mel TregS in exosome, and successfully discriminated between healthy individuals and melanoma patients, as well as early and late-stage patients. This approach significantly enhanced detection sensitivity. In sum, our research elucidated fresh insights into the mechanisms governing Treg self-maintenance and communication with surrounding cells in melanoma. We also introduced an innovative method for clinical disease monitoring through SERS technology.
Collapse
Affiliation(s)
- Tengda Li
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.
| | - Tianqin Wu
- The 100th Hospital of PLA, Suzhou 215006, China
| | - Xiang Li
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Cheng Qian
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China; Department of Laboratory Medicine, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, China.
| |
Collapse
|
48
|
Dong C, Lin JM, Wang Y, Zhu J, Lin L, Xu J, Du J. Exploring the Common Pathogenic Mechanisms of Psoriasis and Atopic Dermatitis: The Interaction between SGK1 and TIGIT Signaling Pathways. Inflammation 2024:10.1007/s10753-024-02115-1. [PMID: 39088121 DOI: 10.1007/s10753-024-02115-1] [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: 05/01/2024] [Revised: 07/17/2024] [Accepted: 07/26/2024] [Indexed: 08/02/2024]
Abstract
This study aims to explore the common pathogenic mechanisms of psoriasis and atopic dermatitis, two T-cell-mediated autoimmune diseases. Utilizing single-cell transcriptomic sequencing data, we revealed that Treg cells primarily express TIGIT in both psoriasis and atopic dermatitis, and identified a subset of macrophages that highly express SGK1. These cells can interact with T cells via the NECTIN2-TIGIT signaling pathway, inhibiting the differentiation of T cells into a pro-inflammatory phenotype, thereby uncovering a common immunoregulatory mechanism in both diseases. Furthermore, we discovered that inhibition of SGK1 exacerbates the inflammatory response in disease models of both conditions. These findings not only provide a new perspective for a common therapeutic strategy for psoriasis and atopic dermatitis but also highlight the importance of considering these molecular interactions in future treatments. Validation of these observations through further qPCR, immunofluorescence, and animal studies has identified potential new targets for the treatment of psoriasis and atopic dermatitis.
Collapse
Affiliation(s)
- Canbin Dong
- Department of Dermatology, Shanghai Institute of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China
| | - Jui-Ming Lin
- Department of Dermatology, Shanghai Institute of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China
| | - Yilun Wang
- Department of Dermatology, Shanghai Institute of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China
| | - Junhao Zhu
- Department of Dermatology, Shanghai Institute of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China
| | - Lanmei Lin
- Department of Dermatology, Shanghai Institute of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China
| | - Jinhua Xu
- Department of Dermatology, Shanghai Institute of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China
| | - Juan Du
- Department of Dermatology, Shanghai Institute of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China.
| |
Collapse
|
49
|
Wang D, Deng X, Wang J, Che S, Ma X, Zhang S, Dong Q, Huang C, Chen J, Shi C, Zhang MR, Hu K, Luo L, Xiao Z. Environmentally responsive hydrogel promotes vascular normalization to enhance STING anti-tumor immunity. J Control Release 2024; 372:403-416. [PMID: 38914207 DOI: 10.1016/j.jconrel.2024.06.052] [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: 05/28/2024] [Accepted: 06/20/2024] [Indexed: 06/26/2024]
Abstract
The immunosuppressive microenvironment of malignant tumors severely hampers the effectiveness of anti-tumor therapy. Moreover, abnormal tumor vasculature interacts with immune cells, forming a vicious cycle that further interferes with anti-tumor immunity and promotes tumor progression. Our pre-basic found excellent anti-tumor effects of c-di-AMP and RRx-001, respectively, and we further explored whether they could be combined synergistically for anti-tumor immunotherapy. We chose to load these two drugs on PVA-TSPBA hydrogel scaffolds that expressly release drugs within the tumor microenvironment by in situ injection. Studies have shown that c-di-AMP activates the STING pathway, enhances immune cell infiltration, and reverses tumor immunosuppression. Meanwhile, RRx-001 releases nitric oxide, which increases oxidative stress injury in tumor cells and promotes apoptosis. Moreover, the combination of the two presented more powerful pro-vascular normalization and reversed tumor immunosuppression than the drug alone. This study demonstrates a new design option for anti-tumor combination therapy and the potential of tumor environmentally responsive hydrogel scaffolds in combination with anti-tumor immunotherapy.
Collapse
Affiliation(s)
- Duo Wang
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Xiujiao Deng
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China; Department of Pharmacy, The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Jinghao Wang
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China; Department of Pharmacy, The Guangzhou Key Laboratory of Basic and Translational Research on Chronic Diseases, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Shuang Che
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Xiaocong Ma
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China; Department of Radiology, The Fifth Affiliated Hospital of Jinan University (Shenhe People's Hospital), Heyuan 517000, China
| | - Siqi Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Qiu Dong
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Cuiqing Huang
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Jifeng Chen
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Changzheng Shi
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China.
| | - Ming-Rong Zhang
- Department of Advanced Nuclear Medicine Sciences, Institute of Quantum Medical, Science, National Institutes for Quantum Science and Technology, Chiba 2638555, Japan
| | - Kuan Hu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China.
| | - Liangping Luo
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China; Department of Radiology, The Fifth Affiliated Hospital of Jinan University (Shenhe People's Hospital), Heyuan 517000, China.
| | - Zeyu Xiao
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou 510632, China.
| |
Collapse
|
50
|
Hua S, Gu X, Jin H, Zhang X, Liu Q, Yang J. Tumor-infiltrating T lymphocytes: A promising immunotherapeutic target for preventing immune escape in cholangiocarcinoma. Biomed Pharmacother 2024; 177:117080. [PMID: 38972151 DOI: 10.1016/j.biopha.2024.117080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/22/2024] [Accepted: 06/29/2024] [Indexed: 07/09/2024] Open
Abstract
Cholangiocarcinoma (CCA) is becoming more common and deadly worldwide. Tumor-infiltrating T cell subtypes make distinct contributions to the immune system; collectively, they constitute a significant portion of the tumor microenvironment (TME) in CCA. By secreting cytokines and other chemicals, regulatory T cells (Tregs) decrease activated T cell responses, acting as immunosuppressors. Reduced CD8+ T cell activation results in stimulating programmed death-1 (PD-1), which undermines the immunological homeostasis of T lymphocytes. On the other hand, cancer cells are eliminated by activated cytotoxic T lymphocyte (CTL) through the perforin-granzyme or Fas-FasL pathways. Th1 and CTL immune cell infiltration into the malignant tumor is also facilitated by γδ T cells. A higher prognosis is typically implied by CD8+ T cell infiltration, and survival is inversely associated with Treg cell density. Immune checkpoint inhibitors, either singly or in combination, provide novel therapeutic strategies for CCA immunotherapy. Furthermore, it is anticipated that immunotherapeutic strategies-such as the identification of new immune targets, combination treatments involving several immune checkpoint inhibitors, and chimeric antigen receptor-T therapies (CAR-T)-will optimize the effectiveness of anti-CCA treatments while reducing adverse effects.
Collapse
Affiliation(s)
- Sijia Hua
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou First People's Hospital, Hangzhou, China.
| | - Xinyi Gu
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou First People's Hospital, Hangzhou, China.
| | - Hangbin Jin
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital. School of Medicine, Westlake University, Hangzhou, Zhejiang, China; Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Xiaofeng Zhang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou First People's Hospital, Hangzhou, China; Department of Gastroenterology, Affiliated Hangzhou First People's Hospital. School of Medicine, Westlake University, Hangzhou, Zhejiang, China; Hangzhou Institute of Digestive Diseases, Hangzhou, Zhejiang, China; Zhejiang Provincial Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research, Hangzhou, Zhejiang 310003, China.
| | - Qiang Liu
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital. School of Medicine, Westlake University, Hangzhou, Zhejiang, China; Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang, China.
| | - Jianfeng Yang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou First People's Hospital, Hangzhou, China; Department of Gastroenterology, Affiliated Hangzhou First People's Hospital. School of Medicine, Westlake University, Hangzhou, Zhejiang, China; Hangzhou Institute of Digestive Diseases, Hangzhou, Zhejiang, China; Zhejiang Provincial Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research, Hangzhou, Zhejiang 310003, China.
| |
Collapse
|