1
|
Tanigawa K, Redmond WL. Current landscape and future prospects of interleukin-2 receptor (IL-2R) agonists in cancer immunotherapy. Oncoimmunology 2025; 14:2452654. [PMID: 39812092 PMCID: PMC11740684 DOI: 10.1080/2162402x.2025.2452654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/07/2025] [Accepted: 01/08/2025] [Indexed: 01/16/2025] Open
Abstract
Immune checkpoint blockade (ICB) has significantly improved the survival for many patients with advanced malignancy. However, fewer than 50% of patients benefit from ICB, highlighting the need for more effective immunotherapy options. High-dose interleukin-2 (HD IL-2) immunotherapy, which is approved for patients with metastatic melanoma and renal cell carcinoma, stimulates CD8+ T cells and NK cells and can generate durable responses in a subset of patients. Moreover, HD IL-2 may have potential efficacy in patients whose disease has progressed following ICB and plays a vital role in expanding tumor-infiltrating lymphocyte (TIL) in TIL therapy. Despite its potential, the use of HD IL-2 is limited by severe toxicities such as hypotension and vascular leak syndrome. Additionally, only a few patients achieve a good outcome after HD IL-2 therapy. To address these challenges, numerous next-generation IL-2 receptor (IL-2 R) agonists have been developed to exhibit treatment effects while minimizing adverse events. This review will explore IL-2 biology, the clinical application of HD IL-2 therapy, and the development of novel IL-2 R agonists for cancer immunotherapy.
Collapse
Affiliation(s)
- Kengo Tanigawa
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
- Department of Pulmonary Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - William L. Redmond
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| |
Collapse
|
2
|
Zhang J, Yuan Y, Ni S, Mu S, Wang W, Sun F, Liang B, Lu P, Qiu Y, Du W, Wang C, Duan H, Hu Z, Wen L, Zheng X, Sheng Y, Zhang S, Chen S, Yin X, Zhu Z. LncRNA PVT1 regulates CD4 + T cell dysregulation in systemic lupus erythematosus: insights from human patients and MRL/lpr mouse. Clin Rheumatol 2025:10.1007/s10067-025-07519-4. [PMID: 40493320 DOI: 10.1007/s10067-025-07519-4] [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: 12/22/2024] [Revised: 04/17/2025] [Accepted: 05/26/2025] [Indexed: 06/12/2025]
Abstract
OBJECTIVES To investigate the role of lncRNA PVT1 in modulating CD4+ T cell subsets and its contribution to systemic lupus erythematosus (SLE) pathogenesis in human patients and MRL/lpr mice. METHODS Measured PVT1 and miR-30e-5p expression in SLE patients (n = 65) and healthy controls (HCs) using qRT-PCR. Analyzed Th1/Th2/Th17/Treg cell frequencies by flow cytometry and cytokine levels (IL-2, IL-4, IL-6, IL-17, TGF-β) via ELISA. Constructed lentiviral vectors to silence (SLE + si-Pvt1) or overexpress Pvt1 (SLE + lenti-Pvt1) in MRL/lpr mice (n = 40). RESULTS PVT1 was upregulated (p = 0.0488) and miR-30e-5p downregulated (p = 0.0095) in SLE patients. Th2 (p = 0.0165) and Th17 (p = 0.0017) cells exhibited a significant increase, while Th1 and Treg cells decreased. Pvt1 silencing reversed SLE phenotypes, increasing Th1 and Treg cells, reducing Th2 and Th17 cells, restoring IL-2 and TGF-β levels and reducing levels of IL-6 and IL-17. Overexpression of Pvt1 exacerbated disease severity. Pvt1 acted as a ceRNA to sponge miR-30e-5p, modulating T-bet/GATA3/RORγt/Foxp3 expression. CONCLUSIONS PVT1 dysregulation disrupts CD4+ T cell homeostasis in SLE. Targeting the PVT1/miR-30e-5p axis may restore immune balance and represent a novel therapeutic strategy. Key Points • Our data confirm the imbalance of CD4+ T cell subsets in SLE patients and demonstrate specific upregulation of lncRNA PVT1 expression in female SLE patients. • Targeting lncRNA PVT1 affects Th1/Th2 and Th17/Treg homeostasis in MRL/lpr mice. • Offers fresh insight into the dysregulation of lymphocyte subsets in SLE.
Collapse
Affiliation(s)
- Jiali Zhang
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Hefei, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China
| | - Ying Yuan
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Hefei, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China
| | - Shuangying Ni
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Hefei, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China
| | - Siqi Mu
- First Clinical Medical College, Anhui Medical University, Hefei, China
| | - Wanrong Wang
- First Clinical Medical College, Anhui Medical University, Hefei, China
| | - Feiyang Sun
- First Clinical Medical College, Anhui Medical University, Hefei, China
| | - Bo Liang
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Hefei, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China
| | - Peng Lu
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Hefei, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China
| | - Yue Qiu
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Hefei, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China
| | - Wenhui Du
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Hefei, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China
| | - Chenjun Wang
- Department of Dermatology, Shannan People's Hospital, Shannan, China
| | - Huijie Duan
- Department of Dermatology, Shannan People's Hospital, Shannan, China
| | - Zejuan Hu
- Department of Dermatology, Shannan People's Hospital, Shannan, China
| | - Leilei Wen
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Hefei, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China
| | - Xiaodong Zheng
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Hefei, China
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China
| | - Yujun Sheng
- Department of Dermatology, Friendship Hospital, Beijing, China
| | - Shengquan Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Shanyu Chen
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xueli Yin
- Functional Experiment Center, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Zhengwei Zhu
- Institute of Dermatology and Department of Dermatology, the First Affiliated Hospital, Anhui Medical University, Hefei, China.
- Key Laboratory of Dermatology (Anhui Medical University), Ministry of Education, Hefei, China.
- Department of Dermatology, Shannan People's Hospital, Shannan, China.
| |
Collapse
|
3
|
Kong F, Chen Y, Liu D, Gao H, Yi Q, Zhang M, Li D. Marvelon suppresses MC38 tumor growth and promotes anti-tumor immunity. Mol Immunol 2025; 182:20-29. [PMID: 40158361 DOI: 10.1016/j.molimm.2025.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 03/15/2025] [Accepted: 03/16/2025] [Indexed: 04/02/2025]
Abstract
Colorectal cancer is a prevalent and deadly malignancy globally, posing an important challenge due to its heterogeneity and treatment resistance. Although oral contraceptives have been shown to reduce the incidence of colorectal cancer, their impact on the anti-tumor effect of CD8+ T cells remains unclear. Here we show that the contraceptive Marvelon plays an important role in anti-MC38 tumor immunity. The contraceptive Marvelon significantly inhibits MC38 tumor growth in vivo. Marvelon treatment promotes IFN-γ expression in CD8+ tumor infiltrating lymphocytes, but shows dispensable impact on their exhausted profile. By further investigating the effects of Marvelon's primary components, Ethinylestradiol and Desogestrel, we reveal that Ethinylestradiol enhances IFN-γ production in Type 1 Cytotoxic T (Tc1) cells and significantly inhibits the viability of MC38 tumor cells, whereas Desogestrel exhibits minimal effects. This study not only redefines the role of oral contraceptives but also provides valuable insights for the development of novel immunotherapeutic strategies.
Collapse
Affiliation(s)
- Fandi Kong
- Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Yongyan Chen
- Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Dantong Liu
- Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Hongying Gao
- Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Qiaoru Yi
- Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Mengjuan Zhang
- Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China.
| | - Dan Li
- Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China.
| |
Collapse
|
4
|
Givian A, Azizan A, Jamshidi A, Mahmoudi M, Farhadi E. Iron metabolism in rheumatic diseases. J Transl Autoimmun 2025; 10:100267. [PMID: 39867458 PMCID: PMC11763848 DOI: 10.1016/j.jtauto.2025.100267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 12/24/2024] [Accepted: 01/03/2025] [Indexed: 01/28/2025] Open
Abstract
Iron is a crucial element for living organism in terms of oxygen transport, hematopoiesis, enzymatic activity, mitochondrial respiratory chain function and also immune system function. The human being has evolved a mechanism to regulate body iron. In some rheumatic diseases such as rheumatoid arthritis (RA), systemic lupus erythematous (SLE), systemic sclerosis (SSc), ankylosing spondylitis (AS), and gout, this balanced iron regulation is impaired. Altered iron homeostasis can contribute to disease progression through ROS production, fibrosis, inflammation, abnormal bone homeostasis, NETosis and cell senescence. In this review, we have focused on the iron metabolism in rheumatic disease and its role in disease progression.
Collapse
Affiliation(s)
- Aliakbar Givian
- Rheumatology Research Center, Tehran University of Medical Science, Tehran, Iran
- Department of Immunology, School of Medicine, Semnan University of Medical Science, Semnan, Iran
| | - Amin Azizan
- Rheumatology Research Center, Tehran University of Medical Science, Tehran, Iran
- Research Center for Chronic Inflammatory Diseases, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmadreza Jamshidi
- Rheumatology Research Center, Tehran University of Medical Science, Tehran, Iran
| | - Mahdi Mahmoudi
- Rheumatology Research Center, Tehran University of Medical Science, Tehran, Iran
- Research Center for Chronic Inflammatory Diseases, Tehran University of Medical Sciences, Tehran, Iran
| | - Elham Farhadi
- Rheumatology Research Center, Tehran University of Medical Science, Tehran, Iran
- Research Center for Chronic Inflammatory Diseases, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
MacLean F, Tsegaye AT, Graham JB, Swarts JL, Vick SC, Potchen NB, Cruz Talavera I, Warrier L, Dubrulle J, Schroeder LK, Saito A, Mar C, Thomas KK, Mack M, Sabo MC, Chohan BH, Ngure K, Mugo NR, Lingappa JR, Lund JM, for the Kinga Study Team. Bacterial vaginosis associates with dysfunctional T cells and altered soluble immune factors in the cervicovaginal tract. J Clin Invest 2025; 135:e184609. [PMID: 40131862 PMCID: PMC12077898 DOI: 10.1172/jci184609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 03/17/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUNDBacterial vaginosis (BV) is a dysbiosis of the vaginal microbiome that is prevalent among reproductive-age females worldwide. Adverse health outcomes associated with BV include an increased risk of sexually acquired HIV, yet the immunological mechanisms underlying this association are not well understood.METHODSTo investigate BV-driven changes to cervicovaginal tract (CVT) and circulating T cell phenotypes, Kinga Study participants with or without BV provided vaginal tract (VT) and ectocervical (CX) tissue biopsies and PBMC samples.RESULTSHigh-parameter flow cytometry revealed an increased frequency of cervical CD4+ conventional T (Tconv) cells expressing CCR5 in BR+ versus BR- women. However, we found no difference in the number of CD3+CD4+CCR5+ cells in the CX or VT of BV+ versus BV- individuals, suggesting that BV-driven increased HIV susceptibility may not be solely attributed to increased CVT HIV target cell abundance. Flow cytometry also revealed that individuals with BV had an increased frequency of dysfunctional CX and VT CD39+ Tconv and CX tissue-resident CD69+CD103+ Tconv cells, reported to be implicated in HIV acquisition risk and replication. Many soluble immune factor differences in the CVT further support that BV elicits diverse and complex CVT immune alterations.CONCLUSIONOur comprehensive analysis expands on potential immunological mechanisms that may underlie the adverse health outcomes associated with BV, including increased HIV susceptibility.TRIAL REGISTRATIONClinicalTrials.gov NCT03701802.FUNDINGThis work was supported by National Institutes of Health grants R01AI131914, R01AI141435, and R01AI129715.
Collapse
Affiliation(s)
- Finn MacLean
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | | | - Jessica B. Graham
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Jessica L. Swarts
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Sarah C. Vick
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Nicole B. Potchen
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Irene Cruz Talavera
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Lakshmi Warrier
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Julien Dubrulle
- Cellular Imaging Shared Resource, Fred Hutchinson Cancer Center. Seattle, Washington, USA
| | - Lena K. Schroeder
- Cellular Imaging Shared Resource, Fred Hutchinson Cancer Center. Seattle, Washington, USA
| | - Ayumi Saito
- Department of Global Health, University of Washington, Seattle, Washington, USA
| | - Corinne Mar
- Department of Global Health, University of Washington, Seattle, Washington, USA
| | - Katherine K. Thomas
- Department of Global Health, University of Washington, Seattle, Washington, USA
| | - Matthias Mack
- Department of Internal Medicine–Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Michelle C. Sabo
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Bhavna H. Chohan
- Department of Global Health, University of Washington, Seattle, Washington, USA
- Center for Virus Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Kenneth Ngure
- Department of Global Health, University of Washington, Seattle, Washington, USA
- School of Public Health, Jomo Kenyatta University of Agriculture and Technology, Nairobi, Kenya
| | - Nelly Rwamba Mugo
- Department of Global Health, University of Washington, Seattle, Washington, USA
- Center for Clinical Research, Kenya Medical Research Institute, Nairobi, Kenya
| | - Jairam R. Lingappa
- Department of Global Health, University of Washington, Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Jennifer M. Lund
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Global Health, University of Washington, Seattle, Washington, USA
| | | |
Collapse
|
6
|
Huang R, Wang Y, Teng H, Xu M, He K, Shen Y, Guo G, Feng X, Li T, Zhou B, Bajenoff M, Lawrence T, Liang Y, Lu L, Zhang L. Tyrosinase in melanoma inhibits anti-tumor activity of PD-1 deficient T cells. BMC Biol 2025; 23:135. [PMID: 40375241 PMCID: PMC12083179 DOI: 10.1186/s12915-025-02237-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 05/06/2025] [Indexed: 05/18/2025] Open
Abstract
BACKGROUND Melanoma is one of the most commonly diagnosed malignancies and serves as a model for studying immunotherapy. The B16 melanoma model, resembling human cold tumors that lack T cell infiltration and show minimal response to PD-1 blockade, is widely used for studying melanoma and its resistance to immunotherapy. Therefore, understanding the molecular basis that prevents T cell-mediated anti-tumor activity in B16 melanoma is of great significance. RESULTS In this study, we generated tyrosinase knockout B16 melanoma cells using CRISPR/Cas9 and discovered that tyrosinase in melanoma significantly inhibits the anti-tumor activity of T cells. Tyrosinase deficiency leads to a 3.80-fold increase in T-cell infiltration and enhances T-cell activation within the tumor. Single-cell RNA sequencing reveals an altered cold tumor immunophenotype in tyrosinase-deficient B16 melanoma. In wild-type mice, T cells in tyrosinase-deficient tumors express elevated levels of PD-1 and Foxp3. However, strikingly, in PD-1 deficient mice, the loss of tyrosinase in B16 melanoma unleashes the anti-tumor activity of PD-1 deficient T cells. This enhanced anti-tumor activity is explained by significantly increased tumor T cell infiltration accompanied by reduced frequencies of regulatory T cells in PD-1 knockout mice. CONCLUSIONS These findings suggest that targeting tyrosinase could convert cold tumors into an immune-responsive state in vivo using murine models. Inhibiting tyrosinase could enhance the effectiveness of PD-1 blockade, offering a new approach for melanoma patients who fail in current PD-1 inhibitor treatment.
Collapse
Affiliation(s)
- Rong Huang
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang, China.
| | - Yingbin Wang
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang, China
| | - Haitao Teng
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang, China
| | - Mengjun Xu
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang, China
| | - Kexin He
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang, China
| | - Yingzhuo Shen
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang, China
| | - Guo Guo
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang, China
- Centre d'immunologie de Marseille-Luminy, Aix-Marseille University, CNRS, INSERM, Marseille, France
| | - Xinyu Feng
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang, China
| | - Tianhan Li
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Binhui Zhou
- Institute of Psychiatry and Neuroscience, Xinxiang Medical University, Xinxiang, China
| | - Marc Bajenoff
- Centre d'immunologie de Marseille-Luminy, Aix-Marseille University, CNRS, INSERM, Marseille, France
| | - Toby Lawrence
- Centre d'immunologie de Marseille-Luminy, Aix-Marseille University, CNRS, INSERM, Marseille, France
- Centre for Inflammation Biology and Cancer Immunology, School of Immunology & Microbial Sciences, King's College London, London, UK
| | - Yinming Liang
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang, China.
- Center of Disease Model and Immunology, Hunan Academy of Chinese Medicine, Changsha, China.
| | - Liaoxun Lu
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang, China.
| | - Lichen Zhang
- Laboratory of Genetic Regulators in the Immune System, School of Medical Technology, Xinxiang Medical University, Xinxiang, China.
- Center of Disease Model and Immunology, Hunan Academy of Chinese Medicine, Changsha, China.
| |
Collapse
|
7
|
Marshall L, Raychaudhuri S, Viatte S. Understanding rheumatic disease through continuous cell state analysis. Nat Rev Rheumatol 2025:10.1038/s41584-025-01253-6. [PMID: 40335652 DOI: 10.1038/s41584-025-01253-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/03/2025] [Indexed: 05/09/2025]
Abstract
Autoimmune rheumatic diseases are a heterogeneous group of conditions, including rheumatoid arthritis (RA) and systemic lupus erythematosus. With the increasing availability of large single-cell datasets, novel disease-associated cell types continue to be identified and characterized at multiple omics layers, for example, 'T peripheral helper' (TPH) (CXCR5- PD-1hi) cells in RA and systemic lupus erythematosus and MerTK+ myeloid cells in RA. Despite efforts to define disease-relevant cell atlases, the very definition of a 'cell type' or 'lineage' has proven controversial as higher resolution assays emerge. This Review explores the cell types and states involved in disease pathogenesis, with a focus on the shifting perspectives on immune and stromal cell taxonomy. These understandings of cell identity are closely related to the computational methods adopted for analysis, with implications for the interpretation of single-cell data. Understanding the underlying cellular architecture of disease is also crucial for therapeutic research as ambiguity hinders translation to the clinical setting. We discuss the implications of different frameworks for cell identity for disease treatment and the discovery of predictive biomarkers for stratified medicine - an unmet clinical need for autoimmune rheumatic diseases.
Collapse
Affiliation(s)
- Lysette Marshall
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester, UK
| | - Soumya Raychaudhuri
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester, UK
- Center for Data Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Divisions of Rheumatology, Inflammation and Immunity and Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute, Cambridge, MA, USA
| | - Sebastien Viatte
- Centre for Genetics and Genomics Versus Arthritis, Centre for Musculoskeletal Research, The University of Manchester, Manchester, UK.
- NIHR Manchester Musculoskeletal Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK.
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.
| |
Collapse
|
8
|
Lin YL, Wang CR. Diet-induced dyslipidemia enhances IFN-γ production in mycolic acid-specific T cells and affects mycobacterial control. Mucosal Immunol 2025:S1933-0219(25)00047-9. [PMID: 40324594 DOI: 10.1016/j.mucimm.2025.04.009] [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: 11/14/2024] [Revised: 03/21/2025] [Accepted: 04/30/2025] [Indexed: 05/07/2025]
Abstract
Dyslipidemia, characterized by altered lipid profiles, influences host immune responses against infections, including Mycobacterium tuberculosis (Mtb). While the effects of dyslipidemia on conventional T cell responses are well documented, its impact on group 1-CD1 restricted T cells, a distinct subset of lipid antigen-specific unconventional T cells, during Mtb infection remains unclear. In this study, we developed a double-transgenic mouse model expressing human group 1 CD1 (hCD1Tg) and mycolic acid (MA)-specific CD1b-restricted T cell receptor (DN1Tg) in a Rag-deficient and low-density lipoprotein receptor-deficient background to investigate how diet-induced dyslipidemia affects the functionality of MA-specific T cells and their role in anti-Mtb immunity. We found that diet-induced dyslipidemia led to increased IFN-γ production by MA-specific T cells, which promoted mycobacterial clearance in vitro. Mechanistically, this enhanced IFN-γ production was associated with increased TCR signaling and enhanced glycolysis in DN1 T cells, rather than changes in antigen presentation by dendritic cells. However, dyslipidemia also increased apoptosis in DN1 T cells, which may have impaired their ability to control mycobacterial infection in vivo, resulting in reduced bacterial clearance. These findings highlight a complex interplay between diet-induced dyslipidemia and lipid antigen-specific T-cell responses in Mtb infection, providing insights for potential therapeutic strategies to mitigate dyslipidemia-induced changes in T-cell functions.
Collapse
Affiliation(s)
- Yen-Lin Lin
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, USA
| | - Chyung-Ru Wang
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, USA.
| |
Collapse
|
9
|
Sadighi Akha AA, Csomós K, Ujházi B, Walter JE, Kumánovics A. Evolving Approach to Clinical Cytometry for Immunodeficiencies and Other Immune Disorders. Immunol Allergy Clin North Am 2025; 45:205-221. [PMID: 40287169 DOI: 10.1016/j.iac.2025.01.009] [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] [Indexed: 04/29/2025]
Abstract
Primary immunodeficiencies were initially identified on the basis of recurrent, severe or unusual infections. Subsequently, it was noted that these diseases can also manifest with autoimmunity, autoinflammation, allergy, lymphoproliferation and malignancy, hence a conceptual change and their renaming as inborn errors of immunity. Ongoing advances in flow cytometry provide the opportunity to expand or modify the utility and scope of existing laboratory tests in this field to mirror this conceptual change. Here we have used the B cell subset, variably known as CD21low B cells, age-associated B cells and T-bet+ B cells, as an example to demonstrate this possibility.
Collapse
Affiliation(s)
- Amir A Sadighi Akha
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Krisztián Csomós
- Division of Pediatric Allergy/Immunology, University of South Florida, Johns Hopkins All Children's Hospital, St. Petersburg, FL, USA
| | - Boglárka Ujházi
- Division of Pediatric Allergy/Immunology, University of South Florida, Johns Hopkins All Children's Hospital, St. Petersburg, FL, USA
| | - Jolán E Walter
- Division of Pediatric Allergy/Immunology, University of South Florida, Johns Hopkins All Children's Hospital, St. Petersburg, FL, USA
| | - Attila Kumánovics
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
10
|
Sala E, Nelli M, Laura C, Di Lucia P, Beccaria CG, Bono EB, Mangione M, Marotta D, Sperto V, Grillo M, Giustini L, Tosi F, Nie J, Kim D, Furiato G, Malpighi C, Consolo E, Becher B, David E, Cohen M, Giladi A, Amit I, Bosselut R, Guidotti LG, Iannacone M, Kuka M. T-cell-derived IFN-γ suppresses T follicular helper cell differentiation and antibody responses. EMBO J 2025; 44:2400-2423. [PMID: 40169810 PMCID: PMC12048687 DOI: 10.1038/s44318-025-00414-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 02/23/2025] [Accepted: 03/03/2025] [Indexed: 04/03/2025] Open
Abstract
CD4+ T cells play a critical role in antiviral humoral and cellular immune responses. We have previously reported that subcutaneous lymphocytic choriomeningitis virus (s.c. LCMV) infection is characterized by a stark compartmentalization of CD4+ T cells, leading to strong TH1 cell polarization but virtually absent T follicular helper (TFH) cells, key drivers of humoral immunity. Here, we investigate the mechanisms responsible for this impaired TFH differentiation. We show that T-bet+ cells induced by LCMV infection encompass a TH1 cell subset expressing granzyme B (GzmB), and a Tcf-1+ cell subset that retains the potential for TFH differentiation without expressing mature TFH markers. Notably, IFN-γ blockade enables full differentiation of Tcf-1+ cells into TFH cells, formation of germinal centers, and increased antibody production. Suppression of TFH cells by IFN-γ is not directly mediated by CD4+ T cells but rather involves another cell type, likely dendritic cells (DCs). Our study provides novel insights into the mechanisms underlying early CD4+ T-cell polarization and humoral responses to viruses, with the potential to facilitate the development of effective vaccine strategies.
Collapse
Affiliation(s)
- Eleonora Sala
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maria Nelli
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Chiara Laura
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Pietro Di Lucia
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Cristian Gabriel Beccaria
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Elisa B Bono
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marta Mangione
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Davide Marotta
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Valentina Sperto
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marta Grillo
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Leonardo Giustini
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Fabio Tosi
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Jia Nie
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Daehong Kim
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Giuliana Furiato
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Chiara Malpighi
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Eleonora Consolo
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Eyal David
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Merav Cohen
- Department of Clinical Microbiology and Immunology, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Amir Giladi
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Ido Amit
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Remy Bosselut
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Luca G Guidotti
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Matteo Iannacone
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy.
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy.
- Experimental Imaging Centre, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| | - Mirela Kuka
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy.
- Division of Immunology, Transplantation, and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
11
|
Hughes EP, Manna AK, Sun W, Osburn-Staker SM, Aamodt S, Warren KJ, Cox JE, Tantin D. Transcriptional co-regulator OCA-B/Pou2af1 restricts Th2 differentiation. Front Immunol 2025; 16:1548636. [PMID: 40364837 PMCID: PMC12069319 DOI: 10.3389/fimmu.2025.1548636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 04/07/2025] [Indexed: 05/15/2025] Open
Abstract
Background Type 2 immunity is initiated through a synergistic response between innate and adaptive immune cells to facilitate host-pathogen defense and wound repair, yet aberrant responses can contribute to chronic inflammation and allergic disease. CD4+ type 2 helper T (Th2) cells facilitate the adaptive immune response through the secretion of cytokines such as IL-4, IL-5, and IL-13. While the Th2 program is governed by the transcription factor GATA3, less is known about regulators that fine-tune the Th2 cytokine response. Method We used a proximity labeling system to map proteins associated with the transcriptional co-regulator OCA-B, encoded by Pou2af1, in T cells. We used a series of genomic, biochemical and immunological assays to probe the interaction with one particular hit from the screen. Results We find that OCA-B indirectly associates with GATA3. ChIP-seq analysis reveals coenrichment of Gata3 and the transcription factor Oct1, a partner protein of OCA-B, at genomic locations responsible for the Th2 program including Il4, Il13, Il5, Gata3, and Irf4. DNA binding data using recombinant proteins and reporter data using T cell lines are consistent with a model in which OCA-B restricts transcription at the Th2 locus control region and subsequent IL-4 and IL-13 secretion. Finally, in an in vivo papain allergy model we show OCA-B expression in T cells limits the frequency of T cells within the lung. Conclusion These findings shown that OCA-B helps restrict Th2 function at least in part through communication with GATA3.
Collapse
Affiliation(s)
- Erik P. Hughes
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, United States
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Asit K. Manna
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, United States
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Wenxiang Sun
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Sandra M. Osburn-Staker
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Samuel Aamodt
- Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Kristi J. Warren
- Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - James E. Cox
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, United States
| | - Dean Tantin
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT, United States
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, UT, United States
| |
Collapse
|
12
|
Paton H, Sarkar P, Gurung P. An overview of host immune responses against Leishmania spp. infections. Hum Mol Genet 2025:ddaf043. [PMID: 40287829 DOI: 10.1093/hmg/ddaf043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 03/18/2025] [Accepted: 03/24/2025] [Indexed: 04/29/2025] Open
Abstract
Leishmania spp. infections pose a significant global health challenge, affecting approximately 1 billion people across more than 88 endemic countries. This unicellular, obligate intracellular parasite causes a spectrum of diseases, ranging from localized cutaneous lesions to systemic visceral infections. Despite advancements in modern medicine and increased understanding of the parasite's etiology and associated diseases, treatment options remain limited to pentavalent antimonials, liposomal amphotericin B, and miltefosine. A deeper understanding of the interactions between immune and non-immune cells involved in the clearance of Leishmania spp. infections could uncover novel therapeutic strategies for this debilitating disease. This review highlights recent progress in elucidating how various cell types contribute to the regulation and resolution of Leishmania spp. infections.
Collapse
Affiliation(s)
- Hanna Paton
- Inflammation Program, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
- Department of Internal Medicine, University of Iowa, 431 Newton Road, Iowa City, IA 52442, United States
- Immunology Graduate Program, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
| | - Prabuddha Sarkar
- Inflammation Program, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
- Department of Internal Medicine, University of Iowa, 431 Newton Road, Iowa City, IA 52442, United States
| | - Prajwal Gurung
- Inflammation Program, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
- Department of Internal Medicine, University of Iowa, 431 Newton Road, Iowa City, IA 52442, United States
- Immunology Graduate Program, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
- Interdisciplinary Graduate Program in Human Toxicology, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
- Center for Immunology and Immune Based Disease, University of Iowa, 431 Newton Road, Iowa City, IA 52242, United States
- Iowa City Veterans Affairs (VA) Medical Center, 601 US-6, Iowa City, IA 52246, United States
| |
Collapse
|
13
|
Kolland D, Kuhlmann M, de Almeida GP, Köhler A, Arifovic A, von Strempel A, Pourjam M, Bolsega S, Wurmser C, Steiger K, Basic M, Neuhaus K, Schmidt-Weber CB, Stecher B, Zehn D, Ohnmacht C. A specific microbial consortium enhances Th1 immunity, improves LCMV viral clearance but aggravates LCMV disease pathology in mice. Nat Commun 2025; 16:3902. [PMID: 40274773 PMCID: PMC12022176 DOI: 10.1038/s41467-025-59073-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: 02/07/2024] [Accepted: 04/10/2025] [Indexed: 04/26/2025] Open
Abstract
Anti-viral immunity can vary tremendously from individual to individual but mechanistic understanding is still scarce. Here, we show that a defined, low complex bacterial community (OMM12) but not the general absence of microbes in germ-free mice leads to a more potent immune response compared to the microbiome of specific-pathogen-free (SPF) mice after a systemic viral infection with LCMV Clone-13. Consequently, gnotobiotic mice colonized with OMM12 have more severe LCMV-induced disease pathology but also enhance viral clearance in the intestinal tract. Mechanistically, single-cell RNA sequencing analysis of adoptively transferred virus-specific T helper cells and endogenous T helper cells in the intestinal tract reveal a stronger pro-inflammatory Th1 profile and a more vigorous expansion in OMM12 than SPF mice. Altogether, our work highlights the causative function of the intestinal microbiome for shaping adaptive anti-viral immunity with implications for vaccination strategies and anti-cancer treatment regimens.
Collapse
Affiliation(s)
- Daphne Kolland
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center, Munich, Germany
| | - Miriam Kuhlmann
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Gustavo P de Almeida
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
- Center for Infection Prevention (ZIP), School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Amelie Köhler
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center, Munich, Germany
| | - Anela Arifovic
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center, Munich, Germany
| | - Alexandra von Strempel
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Faculty of Medicine, LMU, Munich, Germany
| | - Mohsen Pourjam
- Core Facility Microbiome ZIEL - Institute for Food & Health, Technical University of Munich, Freising, Germany
| | - Silvia Bolsega
- Institute for Laboratory Animal Science and Central Animal Facility, Hannover Medical School, Hannover, Germany
| | - Christine Wurmser
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
- Center for Infection Prevention (ZIP), School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Katja Steiger
- Institute of Pathology, School of Medicine and Health, Technical University Munich, Munich, Germany
| | - Marijana Basic
- Institute for Laboratory Animal Science and Central Animal Facility, Hannover Medical School, Hannover, Germany
| | - Klaus Neuhaus
- Core Facility Microbiome ZIEL - Institute for Food & Health, Technical University of Munich, Freising, Germany
| | - Carsten B Schmidt-Weber
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center, Munich, Germany
- Member of the German Center of Lung Research (DZL), Partner Site Munich, Munich, Germany
| | - Bärbel Stecher
- Max von Pettenkofer Institute of Hygiene and Medical Microbiology, Faculty of Medicine, LMU, Munich, Germany
- German Center for Infection Research (DZIF), partner site LMU, Munich, Germany
| | - Dietmar Zehn
- Division of Animal Physiology and Immunology, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany.
- Center for Infection Prevention (ZIP), School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany.
| | - Caspar Ohnmacht
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center, Munich, Germany.
| |
Collapse
|
14
|
Qiu X, Wen R, Wu F, Mao J, Azad T, Wang Y, Zhu J, Zhou X, Xie H, Hong K, Li B, Zhang L, Wen C. The role of double-negative B cells in the pathogenesis of systemic lupus erythematosus. Autoimmun Rev 2025; 24:103821. [PMID: 40274006 DOI: 10.1016/j.autrev.2025.103821] [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: 02/10/2025] [Revised: 04/06/2025] [Accepted: 04/21/2025] [Indexed: 04/26/2025]
Abstract
B cells are essential to the pathophysiology of systemic lupus erythematosus (SLE), a chronic autoimmune illness. IgD-CD27-double negative B cells (DNB cells) are one of the aberrant B cell subsets linked to SLE that have attracted much scientific interest. There is growing evidence that DNB cells play a significant role in the development of the disease and are strongly linked to the activity of lupus. These cells play a pivotal role in the pathogenesis of SLE by producing a diverse array of autoantibodies, which form immune complexes that drive target organ damage. A comprehensive understanding of SLE pathophysiology necessitates in-depth investigation into DNB cells, not only to elucidate their mechanistic contributions but also to uncover novel therapeutic strategies. According to available data, treatments that target B cells have proven effective in managing SLE; nevertheless, a significant breakthrough in precision medicine for SLE may come from targeting DNB cells specifically. Despite growing interest in DNB cells, their precise characteristics, developmental trajectories, and regulatory mechanisms remain incompletely defined, posing significant challenges to the field. A comprehensive investigation of the regulatory mechanisms governing DNB cell differentiation and expansion in SLE may facilitate novel therapeutic discoveries. This review aims to provide an updated synthesis of current research on DNB cells, with a focus on their origins, developmental trajectories in SLE, and potential as precision therapeutic targets.
Collapse
Affiliation(s)
- Xinying Qiu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 410011 Changsha, China; The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University, Changsha 410007, Hunan, China
| | - RuiFan Wen
- Medical School, Hunan University of Chinese Medicine, No.300 Xueshi Road, Hanpu Science & Education District, Changsha, Hunan 410208, China
| | - Feifeng Wu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Jueyi Mao
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Tasnim Azad
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Yang Wang
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Junquan Zhu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Xin Zhou
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Haotian Xie
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Kimsor Hong
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Binbin Li
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Liang Zhang
- The Affiliated Children's Hospital of Xiangya School of Medicine, Central South University, Changsha 410007, Hunan, China; Department of Nephrology, Rheumatology and Immunology, Hunan Children's Hospital, Changsha 410007, Hunan, China.
| | - Chuan Wen
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, 410011 Changsha, China.
| |
Collapse
|
15
|
Cimons JM, DeGolier KR, Burciaga SD, Yarnell MC, Novak AJ, Rivera-Reyes AM, Kohler ME, Fry TJ. T-bet overexpression enhances CAR T cell effector functions and antigen sensitivity. J Immunother Cancer 2025; 13:e010962. [PMID: 40246581 PMCID: PMC12007057 DOI: 10.1136/jitc-2024-010962] [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: 11/01/2024] [Accepted: 04/07/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND T cells modified to express a chimeric antigen receptor (CAR) are successful against B-lineage malignancies but fail to induce durable remissions in up to half of patients and have shown limited efficacy against other types of cancer. Strategies to improve CAR T cell potency and responses to low antigen densities without inducing CAR T cell dysfunction or limiting persistence are necessary to expand durability of remissions. METHODS We overexpressed T-bet in human and mouse CAR T cells to mimic exposure to signal 3 cytokines during T cell priming to promote T helper cell 1 (Th1) polarization of CD4+CAR T cells with the goal of enhancing antitumor activity. Using human CAR T cells and xenograft models we interrogated the impact of T-bet overexpression on CAR T cell antitumor activity in vitro and in vivo. We also used a syngeneic murine CAR T cell model to study the impact of T-bet overexpression on long-term persistence and secondary responses to tumor rechallenge. RESULTS T-bet overexpression reduced expression of the Th2 cytokine interleukin 4 and promoted polyfunctional production of Th1-associated cytokines in response to CAR stimulation. T-bet overexpression enhanced some effector functions in vitro but did not improve CAR T cell-mediated control of leukemia expressing high levels of antigen in vivo. T-bet overexpression also improved effector function of murine CD19 CAR T cells with no impairment to the persistence or ability of persistent CAR T cells to re-expand and clear a secondary leukemia challenge. Finally, T-bet overexpression promoted enhanced in vitro function against leukemia expressing low levels of CD19, which translated to improved control of CD19lo leukemia in vivo by human C19 CAR T cells containing a 4-1BB costimulatory domain. CONCLUSIONS Together, our data demonstrate that T-bet overexpression induces a reduction in Th2 cytokine production, an increase in polyfunctional Th1 cytokine production and enhances 4-1BB CAR T cell activity against cancers expressing low levels of target antigen without promoting a loss in functional CAR T cell persistence.
Collapse
Affiliation(s)
- Jennifer M Cimons
- Pediatrics Hematolgy/Oncology/Bone Marrow Transplant, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, Colorado, USA
| | - Kole R DeGolier
- Pediatrics Hematolgy/Oncology/Bone Marrow Transplant, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, Colorado, USA
| | - Samuel D Burciaga
- Pediatrics Hematolgy/Oncology/Bone Marrow Transplant, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, Colorado, USA
| | - Michael C Yarnell
- Pediatrics Hematolgy/Oncology/Bone Marrow Transplant, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, Colorado, USA
| | - Amanda J Novak
- Pediatrics Hematolgy/Oncology/Bone Marrow Transplant, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, Colorado, USA
| | - Amalia M Rivera-Reyes
- Pediatrics Hematolgy/Oncology/Bone Marrow Transplant, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, Colorado, USA
| | - M Eric Kohler
- Pediatrics Hematolgy/Oncology/Bone Marrow Transplant, University of Colorado Anschutz Medical Campus School of Medicine, Aurora, Colorado, USA
- Pediatrics, Children's Hospital Colorado, Aurora, Colorado, USA
| | - Terry J Fry
- University of Colorado Denver Children's Hospital Colorado Research Institute, Aurora, Colorado, USA
| |
Collapse
|
16
|
Wang HL, Yu CX, Yu XM, Lin JJ, Chen YZ, Zhu L. Differential Control of T-Cell Subsets by Recombinant Human PLD2 in a Mouse Model of Allergic Asthma. Immunol Invest 2025; 54:334-351. [PMID: 39692536 DOI: 10.1080/08820139.2024.2441468] [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] [Indexed: 12/19/2024]
Abstract
BACKGROUND Phospholipase D2 (PLD2) enzymes are expressed on the cytoplasmic membrane of bacteria, fungi, plants, and animals. Recently, extensive research has linked PLD2 to the chronic inflammatory activity of cells. Allergic asthma is a chronic airway inflammation disease. In this context, a recombinant human phospholipase D2 (rhPLD2) was designed and modified from the wild-type PLD2 to study its effects in an ovalbumin (OVA) induced murine model of asthma. METHODS Hematoxylin and eosin staining was used for lung histopathology. Cytokine concentrations in bronchoalveolar lavage fluid (BALF) were measured using ELISA kits. The ratio of T-bet and GATA-3 expression level in spleen and lymph nodes following rhPLD2 administration was assessed through RT-PCR. Phenotyping analysis of Treg cells from peripheral blood was performed by flow cytometry. RESULTS It indicated that OVA-induced mice exhibited elevated pulmonary eosinophilia and allergic inflammation in the airways, along with increased expression of IFN-γ, IL-4 in the lung BALF. Administration of rhPLD2 alleviated lung inflammation and significantly reduce the number of eosinophils in peripheral blood and BALF. RhPLD2 also reversed the IFN-γ/IL-4 ratio at the molecular level in BALF and the T-bet/GATA-3 ratio in lymphocytes of the lung, spleen, lymph nodes at the genetic level. Furthermore, FACS analysis demonstrated that rhPLD2 increased the frequency of both IL-10+Treg cells and CD25+ Treg cells. CONCLUSION From a therapeutic perspective, rhPLD2 alleviates allergic airway inflammation by balancing Th1/Th2 homeostasis and increasing Treg cells. It has been shown to function in immunoregulatory activities in OVA-induced asthma mice.
Collapse
Affiliation(s)
- Hui-Li Wang
- Immunology Department and Center of Neuroscience, Fujian Medical University, Fuzhou, Fujian, PR China
| | - Chuan-Xing Yu
- Internal medicine, Second People's Hospital of Fujian Province, Fuzhou, Fujian, PR China
| | - Xiu-Ming Yu
- Immunology Department and Center of Neuroscience, Fujian Medical University, Fuzhou, Fujian, PR China
| | - Jun-Jin Lin
- Immunology Department and Center of Neuroscience, Fujian Medical University, Fuzhou, Fujian, PR China
| | - Yi-Zhong Chen
- Immunology Department and Center of Neuroscience, Fujian Medical University, Fuzhou, Fujian, PR China
| | - Ling Zhu
- Immunology Department and Center of Neuroscience, Fujian Medical University, Fuzhou, Fujian, PR China
| |
Collapse
|
17
|
Devine KJ, Schwartz L, El-Mallawany NK. Peripheral T-cell lymphoma-NOS in children and adolescents: a review from the Children's Oncology Group NHL Committee. Blood Adv 2025; 9:1420-1431. [PMID: 39825825 PMCID: PMC11960635 DOI: 10.1182/bloodadvances.2024013689] [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: 09/20/2024] [Revised: 11/25/2024] [Accepted: 01/11/2025] [Indexed: 01/20/2025] Open
Abstract
ABSTRACT Peripheral T-cell lymphoma not otherwise specified (PTCL-NOS) is a rare mature T-cell non-Hodgkin lymphoma (NHL) seen in both children and adults. Although it is the most common nonanaplastic mature T-cell lymphoma of childhood, it is quite rare and, therefore, the standard of care remains largely undefined. It is a disease characterized by clinical and pathological heterogeneity and is generally associated with an aggressive clinical course and poor prognosis in adults. Retrospective reports on treatment outcomes for pediatric PTCL-NOS are limited by small cohorts, variable clinical presentations, and heterogeneous treatment regimens. Although published survival rates in children appear encouraging compared with those from prospective studies in adults, the prognosis is guarded, and relatively low curative outcomes are in stark contrast to more common pediatric NHL. Although recent landmark gene profiling studies have shed light on the molecular landscape of the disease in adults, identifying molecular subgroups with prognostic significance, the biology of PTCL-NOS remains unclear in children. Here, we review the clinical presentation and diagnosis, historical treatment approaches, current knowledge of the disease biology, and the role of hematopoietic stem cell transplant (HSCT) in PTCL-NOS in children to pursue a better understanding of this heterogeneous condition and empower physicians to use this information to best support our pediatric population. Studies focusing on pediatric PTCL-NOS are required to unravel the disease biology in children, improve risk stratification, and better define upfront treatment through the role of targeted agents and HSCT, as we look to future directions of the care of children with PTCL-NOS.
Collapse
Affiliation(s)
- Kaitlin J. Devine
- Division of Oncology, Children’s Hospital of Philadelphia, Philadelphia, PA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Lindsay Schwartz
- Division of Pediatric Hematology/Oncology, Department of Pediatrics, The University of Chicago, Chicago, IL
| | - Nader Kim El-Mallawany
- Division of Hematology and Oncology, Department of Pediatrics, Baylor College of Medicine, Houston, TX
| |
Collapse
|
18
|
Yan R, Jia D, Qi Y, Wang Q, Chen S. Intestinal tissue-resident memory T cells: Characteristics, functions under physiological and pathological conditions and spatial specificity. J Adv Res 2025:S2090-1232(25)00181-X. [PMID: 40096943 DOI: 10.1016/j.jare.2025.03.021] [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: 01/04/2025] [Revised: 03/08/2025] [Accepted: 03/14/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Tissue-resident memory T (TRM) cells are a distinct subset of memory T cells that persist in non-lymphoid tissues, providing localized and rapid immune responses to infection and malignancy. Unlike circulating memory T cells, TRM cells have unique homing and functional characteristics that are shaped by the tissue microenvironment. In the gut, TRM cells play a pivotal role in maintaining mucosal immunity, exhibiting phenotypic and functional heterogeneity in different intestinal compartments and in response to aging and pathological conditions. AIM OF REVIEW This review aims to systematically examine the definition, spatial heterogeneity and functional roles of intestinal TRM (iTRM) cells. It highlights their contributions to physiological immunity, their involvement in pathological processes such as inflammatory bowel disease (IBD) and colorectal cancer (CRC), and their age-related dynamics. The review also explores emerging therapeutic implications of modulating iTRM cells for intestinal health and disease management. KEY SCIENTIFIC CONCEPTS OF REVIEW: iTRM cells are defined by surface markers like CD69 and CD103, transcriptional regulators such as Hobit, Runx3, Blimp-1, as well as cytokine signals including TGF-β, IFN-β, IL-12. They exhibit spatial and functional heterogeneity across intestinal layers (epithelium versus lamina propria) and regions (small intestine versus colon). In IBD, iTRM cells play a dual role, contributing to both inflammation and tissue repair, whereas in CRC, specific subsets of iTRM cells (e.g., CD8+ CD103+ CD39+) are associated with enhanced antitumor immunity. Aging impacts iTRM functionality, with shifts in the CD4+/CD8+ ratio and reduced cytokine production in elderly individuals. Insights into the metabolic, transcriptional, and environmental regulation of iTRM cells provide avenues for targeted therapies in intestinal diseases, cancer immunotherapy, and interventions to delay intestinal aging.
Collapse
Affiliation(s)
- Ruochen Yan
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, Zhejiang Province 310058, China; Institution of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang Province 310058, China
| | - Dingjiacheng Jia
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310058, China; Institution of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang Province 310058, China
| | - Yadong Qi
- Department of Gastroenterology, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang Province 310058, China; Institution of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang Province 310058, China
| | - Qiwen Wang
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, Zhejiang Province 310058, China; Institution of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang Province 310058, China
| | - Shujie Chen
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, Zhejiang Province 310058, China; Institution of Gastroenterology, Zhejiang University, Hangzhou, Zhejiang Province 310058, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang Province 310001, China.
| |
Collapse
|
19
|
Jablonka-Shariff A, Broberg C, Snyder-Warwick AK. Absence of T-box transcription factor 21 limits neuromuscular junction recovery after nerve injury in T-bet-knockout mice. Front Cell Dev Biol 2025; 13:1535323. [PMID: 40162097 PMCID: PMC11949913 DOI: 10.3389/fcell.2025.1535323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 02/20/2025] [Indexed: 04/02/2025] Open
Abstract
Introduction Terminal Schwann cells (tSCs), at the neuromuscular junction (NMJ), play critical roles in the repair of motor axon terminals at muscle, and rebuild neuronal signaling following nerve injury. Knowledge of mediators impacting tSCs post-nerve injury and in disease may guide beneficial therapies to improve motor outcomes. We previously found T-box transcription factor 21 (TBX21/TBET), classically associated with T-helper1 cells and immune cell recruitment, is expressed in tSCs at the mouse NMJ. The purpose of this study was to examine effects of Tbx21 absence during NMJ regeneration following peripheral nerve injury. Methods Wildtype (WT) and Tbet-knockout (Tbet-KO) mice underwent sciatic nerve transection and immediate repair. Functional muscle recovery assessment was performed with muscle force testing on mice at 2-, 3-, 4-, and 6-week (wks) and 6 months after nerve injury repair. Morphometric analyses of NMJ reinnervation, tSC number, and tSC processes were evaluated. Full NMJ reinnervation was defined as ≥75% coverage of endplates by axons. A minimum of three mice were evaluated in each group, and 50-100 NMJs were evaluated per mouse. Results Tbet-KO mice had significantly diminished muscle function compared to WT mice at every time point beyond 3 weeks. Tbet-KO mice showed just over half of the muscle force generated by WT mice at 4 weeks and 6 weeks post-injury and repair. By 6 months, Tbet-KO mice generated only 84.1% the muscle force of WT mice. Tbet-KO mice showed significantly decreased levels of fully reinnervated NMJs compared to WT mice at each time point tested. Tbet-KO mice also showed a lower number of tSCs with reduced cytoplasmic processes beyond NMJ area and lower number of immune cells during process of NMJ regeneration. Discussion Our findings show that the Tbx21 transcription factor promotes NMJ reinnervation to regain muscle function following nerve injury.
Collapse
Affiliation(s)
- Albina Jablonka-Shariff
- Research Scientist, Division of Plastic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, United States
| | - Curtis Broberg
- Research Student, Washington University School of Medicine, St. Louis, MO, United States
| | - Alison K. Snyder-Warwick
- Division of Plastic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
20
|
Wen Z, Liu T, Xu X, Acharya N, Shen Z, Lu Y, Xu J, Guo K, Shen S, Zhao Y, Wang P, Li S, Chen W, Li H, Ding Y, Shang M, Guo H, Hou Y, Cui B, Shen M, Huang Y, Pan T, Qingqing W, Cao Q, Wang K, Xiao P. Interleukin-16 enhances anti-tumor immune responses by establishing a Th1 cell-macrophage crosstalk through reprogramming glutamine metabolism in mice. Nat Commun 2025; 16:2362. [PMID: 40064918 PMCID: PMC11893787 DOI: 10.1038/s41467-025-57603-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
Overcoming immunosuppression in the tumor microenvironment (TME) is crucial for developing novel cancer immunotherapies. Here, we report that IL-16 administration enhances the polarization of T helper 1 (Th1) cells by inhibiting glutamine catabolism through the downregulation of glutaminase in CD4+ T cells and increases the production of Th1 effector cytokine IFN-γ, thus improving anti-tumor immune responses. Moreover, we find that establishing an IL-16-dependent, Th1-dominant TME relies on mast cell-produced histamine and results in the increased expression of the CXCR3 ligands in tumor-associated macrophages (TAM), thereby improving the therapeutic effectiveness of immune checkpoint blockade (ICB). Cancer patients exhibit impaired production of IL-16, which correlates with poorer prognosis. Additionally, low IL-16 production is associated with unresponsiveness to immunotherapy in cancer patients. Collectively, our findings provided new insights into the biological function of IL-16, emphasizing its potential clinical significance as a therapeutic approach to augment anti-tumor immunity and sensitize ICB-based cancer immunotherapy.
Collapse
Affiliation(s)
- Zhenzhen Wen
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tong Liu
- Department of Breast Surgery, Cancer Hospital of Harbin Medical University, Harbin, China
| | - Xutao Xu
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Nandini Acharya
- Pelotonia Institute for Immuno-Oncology, OSUCCC-James, The Ohio State University, Columbus, OH, USA
| | - Zhida Shen
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yunkun Lu
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Junjie Xu
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ke Guo
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shuying Shen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuening Zhao
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Pinli Wang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Shumin Li
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Weiyu Chen
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China
| | - Hui Li
- Department of Thoracic Medical Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Yimin Ding
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Min Shang
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Hongshan Guo
- Liangzhu Laboratory, Zhejiang University, Hangzhou, China
| | - Yu Hou
- Liangzhu Laboratory, Zhejiang University, Hangzhou, China
| | - Bijun Cui
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Manlu Shen
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Youling Huang
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ting Pan
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
- The Key Laboratory for Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou, China
| | - Wang Qingqing
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China.
- The Key Laboratory for Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou, China.
| | - Qian Cao
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Kai Wang
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, China.
| | - Peng Xiao
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China.
- The Key Laboratory for Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
21
|
Ji Y, Xiao C, Fan T, Deng Z, Wang D, Cai W, Li J, Liao T, Li C, He J. The epigenetic hallmarks of immune cells in cancer. Mol Cancer 2025; 24:66. [PMID: 40038722 PMCID: PMC11881328 DOI: 10.1186/s12943-025-02255-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 01/30/2025] [Indexed: 03/06/2025] Open
Abstract
Targeting the dysregulation of epigenetic mechanisms in cancer has emerged as a promising therapeutic strategy. Although the significant rationale progress of epigenetic therapies in blocking cancer cells, how epigenetic regulation shapes tumor microenvironment (TME) and establishes antitumor immunity remains less understood. Recent study focus has been put on the epigenetic-mediated changes in the fate of immune cells, including the differentiation, expansion, recruitment, functionalization, and exhaustion of T cells, natural killer (NK) cells, tumor-associated macrophages (TAMs), dendritic cells (DCs), myeloid-derived suppressor cells (MDSCs), and B cells within the TME. Here, we review the latest molecular and clinical insights into how DNA modifications, histone modification, and epitranscriptome-related regulations shape immune cells of various cancers. We also discuss opportunities for leveraging epigenetic therapies to improve cancer immunotherapies. This review provides the epigenetic foundations of cancer immunity and proposes the future direction of combination therapies.
Collapse
Affiliation(s)
- Yu Ji
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Chu Xiao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Tao Fan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Ziqin Deng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Di Wang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Wenpeng Cai
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jia Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Tianle Liao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chunxiang Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
22
|
Winslow GM, Levack R. Know Your ABCs: Discovery, Differentiation, and Targeting of T-Bet+ B Cells. Immunol Rev 2025; 330:e13440. [PMID: 39844597 PMCID: PMC11754996 DOI: 10.1111/imr.13440] [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: 11/26/2024] [Revised: 12/22/2024] [Accepted: 01/01/2025] [Indexed: 01/24/2025]
Abstract
Since their first description in 2008, T-bet+ B cells have emerged as a clinically important B cell subset. Now commonly known as ABCs (Age-associated B Cells), they are uniquely characterized by their expression of the transcription factor T-bet. Indeed, this singular factor defines this B cell subset. This review will describe the discovery of T-bet+ B cells, their role in bacterial infection as T cell-independent (TI) plasmablasts, as well as long-term follicular helper T cell-dependent (TD) IgM+ and switched memory cells (i.e., T-bet+ ABCs), and later discoveries of their role(s) in diverse immunological responses. These studies highlight a critical, although limited, role of T-bet in IgG2a class switching, a function central to the cells' role in immunity and autoimmunity. Given their association with autoimmunity, pharmacological targeting is an attractive strategy for reducing or eliminating the B cells. T-bet+ ABCs express a number of characteristic cell surface markers, including CD11c, CD11b, CD73, and the adenosine 2a receptor (A2aR). Accordingly, A2aR agonist administration effectively targeted T-bet+ ABCs in vivo. Moreover, agonist treatment of lupus-prone mice reduced autoantibodies and disease symptoms. This latter work highlights the potential therapeutic use of adenosine agonists for treating autoimmune diseases involving T-bet+ ABCs.
Collapse
Affiliation(s)
- Gary M. Winslow
- Department of Microbiology and ImmunologyUpstate Medical UniversitySyracuseNew YorkUSA
| | - Russell Levack
- Department of ImmunologyUniversity of PittsburghPittsburghPennsylvaniaUSA
| |
Collapse
|
23
|
Zhong L, Wang YH, Kahlfuss S, Jishage M, McDermott M, Yang J, Tao AY, Hu K, Noyer L, Raphael D, Patel D, Knight TE, Chitlur M, Machaca K, Feske S. STIM1-mediated NFAT signaling synergizes with STAT1 to control T-bet expression and T H1 differentiation. Nat Immunol 2025; 26:484-496. [PMID: 39984734 PMCID: PMC12121662 DOI: 10.1038/s41590-025-02089-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 01/13/2025] [Indexed: 02/23/2025]
Abstract
Stromal interaction molecule 1 (STIM1) is critical for store-operated Ca2+ entry (SOCE) and T cell activation. T helper 1 (TH1) cells, which express T-bet (encoded by TBX21), mediate immunity to intracellular pathogens. Although SOCE is known to regulate other TH lineages, its role in Th1 differentiation remains unclear. Here, we report a patient with an intronic loss-of-function mutation in STIM1, which abolishes SOCE and causes immunodeficiency. We demonstrate that SOCE promotes nuclear factor of activated T cells (NFAT) binding to conserved noncoding sequence (CNS)-12 in the TBX21 enhancer and enables NFAT to synergize with STAT1 to mediate TBX21 expression. While SOCE-deficient CD4+ T cells have reduced expression of TBX21 in the absence of interleukin-12 (IL-12), their expression of IL-12 receptors β1 and β2 is increased, sensitizing them to IL-12 signaling and allowing IL-12 to rescue T-bet expression. Our study reveals that the STIM1-SOCE-NFAT signaling axis is essential for the differentiation of Th1 cells depending on the cytokine milieu.
Collapse
Affiliation(s)
- Li Zhong
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Yin-Hu Wang
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Sascha Kahlfuss
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
- Institute of Molecular and Clinical Immunology, Institute of Medical Microbiology and Hospital Hygiene, Medical Faculty, Otto-von-Guericke-University, Magdeburg, Germany
| | - Miki Jishage
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Maxwell McDermott
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Jun Yang
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Anthony Y Tao
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Ke Hu
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Lucile Noyer
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Dimitrius Raphael
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Devisha Patel
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA
| | - Tristan E Knight
- Division of Pediatric Hematology/Oncology, Children's Hospital of Michigan, Detroit, MI, USA
- Kapiolani Medical Center for Women and Children, Burns School of Medicine, Honolulu, HI, USA
| | - Meera Chitlur
- Division of Pediatric Hematology/Oncology, Children's Hospital of Michigan, Detroit, MI, USA
- Central Michigan University College of Medicine, Detroit, MI, USA
| | - Khaled Machaca
- Calcium Signaling Group, Research Department, Weill Cornell Medicine, Doha, Qatar; Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Stefan Feske
- Department of Pathology, New York University Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
24
|
Kotlyar J, Granstein RD. Neuroimmunology of psoriasis: Possible roles for calcitonin gene-related peptide in its pathogenesis. Brain Behav Immun Health 2025; 44:100958. [PMID: 40008232 PMCID: PMC11851231 DOI: 10.1016/j.bbih.2025.100958] [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: 09/24/2024] [Revised: 01/21/2025] [Accepted: 01/29/2025] [Indexed: 02/27/2025] Open
Abstract
The nervous system has a complex interplay with the immune system, especially at barrier sites such as the skin. This allows it to play a role in a variety of cutaneous inflammatory disorders such as psoriasis, exerting effects on various immune cells via effector molecules such as neuropeptides. In this review, we discuss the role of calcitonin gene-related peptide in modulating the immune system and inflammation, with a focus on psoriasis.
Collapse
Affiliation(s)
- Joshua Kotlyar
- Israel Englander Department of Dermatology, Weill Cornell Medicine, 1305 York Avenue, WGC9, New York, NY, 10021, USA
- SUNY Downstate Health Sciences University College of Medicine, 450 Clarkson Avenue, Brooklyn, NY, 11203, USA
| | - Richard D. Granstein
- Israel Englander Department of Dermatology, Weill Cornell Medicine, 1305 York Avenue, WGC9, New York, NY, 10021, USA
| |
Collapse
|
25
|
Xu Z, Zhang Y, Wang Y, Wu A, Meng C, Li W, Yi J, Chen C. Evaluation of the safety and immune protection of OMPAC, PAPF, and EBPSs recombinant subunit vaccines Developed for Escherichia coli, Staphylococcus aureus, and Streptococcus agalactiae in mice. Int Immunopharmacol 2025; 148:114151. [PMID: 39874844 DOI: 10.1016/j.intimp.2025.114151] [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: 07/12/2024] [Revised: 10/30/2024] [Accepted: 01/21/2025] [Indexed: 01/30/2025]
Abstract
Bacterial mastitis in dairy cow is often caused by a combination of bacterial infections, such as Escherichia coli, Staphylococcus aureus, and Streptococcus agalactiae. Currently, there is no effective vaccine against the disease. Therefore, we constructed a recombinant subunit vaccine by fusing gene fragments of E. coli OMPA and OMPC, S. aureus EBPS, and S. agalactiae PGK, AP1, AP2, and FBSA. These gene fragments were combined into three fusion proteins: OMPAC, EBPSs, and PAPF. Mice were immunized with the three fusion proteins either alone or in combination. The test results showed that immunization with OMPAC, EBPSs, and PAPF individually or in combination could induce high titers of antibodies in the mice. Additionally, 21 days post-immunization, IFN-γ levels were significantly increased in all groups of mice, suggesting that immunization with OMPAC, EBPSs, and PAPF, whether alone or in combination, was effective in inducing antibody production. This indicates that OMPAC, EBPSs, and PAPF were effective in inducing both humoral and cellular immunity in mice. Furthermore, immunization with OMPAC, EBPSs, and PAPF individually or in combination were effective in protecting mice from E. coli, S. aureus, and S. agalactiae infections. Importantly, a mixture of the three fusion proteins was relatively safe for pregnant female mice. In conclusion, we successfully constructed and expressed recombinant subunit vaccines of OMPAC, EBPSs and PAPF and verified that these vaccines rapidly induced high levels of specific antibodies while reducing bacterial loads in the organs of mice. This lays the theoretical foundation and data support for the development of novel subunit vaccines against mastitis in dairy cows.
Collapse
Affiliation(s)
- Zhenyu Xu
- College of Animal Science and Technology, Shihezi University 832000 Shihezi City, Xinjiang, China
| | - Yuchen Zhang
- College of Animal Science and Technology, Shihezi University 832000 Shihezi City, Xinjiang, China
| | - Yueli Wang
- College of Animal Science and Technology, Shihezi University 832000 Shihezi City, Xinjiang, China
| | - Aodi Wu
- College of Animal Science and Technology, Shihezi University 832000 Shihezi City, Xinjiang, China
| | - Chuang Meng
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, China
| | - Wei Li
- Xinjiang Center for Animal Disease Control and Prevention, Urumqi, China
| | - Jihai Yi
- College of Animal Science and Technology, Shihezi University 832000 Shihezi City, Xinjiang, China.
| | - Chuangfu Chen
- College of Animal Science and Technology, Shihezi University 832000 Shihezi City, Xinjiang, China.
| |
Collapse
|
26
|
Liao F, Zhou D, Cano M, Liu Z, Scozzi D, Tague LK, Byers DE, Li W, Sivapackiam J, Sharma V, Krupnick AS, Frank DW, Kreisel D, Kulkarni HS, Hachem RR, Gelman AE. Pseudomonas aeruginosa infection induces intragraft lymphocytotoxicity that triggers lung transplant antibody-mediated rejection. Sci Transl Med 2025; 17:eadp1349. [PMID: 39908350 DOI: 10.1126/scitranslmed.adp1349] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 01/16/2025] [Indexed: 02/07/2025]
Abstract
How pathogens inhibit transplant tolerance remains unclear. Here, we found that Pseudomonas aeruginosa infection, but not other common bacterial respiratory infections, increases antibody-mediated rejection (AMR) risk in recipients of lung transplants. To explore this relationship, we performed orthotopic lung transplants in mice, infected recipients with P. aeruginosa, and observed for the development of AMR. Intravital two-photon microscopy showed that P. aeruginosa rapidly invaded bronchial-associated lymphoid tissues, which resulted in acute lymphocytotoxicity, including the death of forkhead box P3 (Foxp3)+CD4+ T cells that are required to suppress AMR. P. aeruginosa-mediated AMR required expression of the type III secretion system (T3SS), which injects exotoxins into the cell cytoplasm. Through a combination of mutagenesis and epitope tagging experiments, we revealed that T3SS exotoxin T ADP ribosyl-transferase activity was sufficient for graft-resident Foxp3+CD4+ T cell apoptosis, leading to myeloid differentiation primary response 88 (Myd88)-dependent generation of T-box expressed in T cells (T-bet)- and C-X-C motif chemokine receptor 3 (CXCR3)-positive germinal center and memory B cells with high donor antigen avidity. We also found that T-bet+ and CXCR3+ B cells were elevated in biopsies from recipients of lung transplants who were diagnosed with AMR. In mice, CXCR3 deficiency restricted to B cells or CXCR3 blockade prevented AMR despite P. aeruginosa infection. Our work has identified a previously unrecognized role of bacterial virulence in lung allograft rejection and suggests potential strategies to prevent AMR for those at high risk of P. aeruginosa infection after transplant.
Collapse
Affiliation(s)
- Fuyi Liao
- Department of Surgery, Division of Cardiothoracic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Dequan Zhou
- Department of Surgery, Division of Cardiothoracic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Marlene Cano
- Department of Medicine, Division of Pulmonology & Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Zhiyi Liu
- Department of Surgery, Division of Cardiothoracic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Davide Scozzi
- Department of Surgery, Division of Cardiothoracic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Laneshia K Tague
- Department of Medicine, Division of Pulmonology & Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Derek E Byers
- Department of Medicine, Division of Pulmonology & Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Wenjun Li
- Department of Surgery, Division of Cardiothoracic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jothilingam Sivapackiam
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Vijay Sharma
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Alexander S Krupnick
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Dara W Frank
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Daniel Kreisel
- Department of Surgery, Division of Cardiothoracic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Hrishikesh S Kulkarni
- Department of Medicine, Division of Pulmonology & Critical Care Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ramsey R Hachem
- Department of Internal Medicine, Division of Respiratory, Critical Care, and Occupational Pulmonary Medicine, University of Utah, Salt Lake City, UT 84108, USA
| | - Andrew E Gelman
- Department of Surgery, Division of Cardiothoracic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
27
|
Johnson SD, Pino M, Acharya A, Clain JA, Bose D, Nguyen K, Harper J, Villinger F, Paiardini M, Byrareddy SN. IL-21 and anti-α4β7 dual therapy during ART promotes immunological and microbiome responses in SIV-infected macaques. JCI Insight 2025; 10:e184491. [PMID: 39903521 PMCID: PMC11949015 DOI: 10.1172/jci.insight.184491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 01/28/2025] [Indexed: 02/06/2025] Open
Abstract
Despite combination antiretroviral therapy (ART), HIV causes persistent gut barrier dysfunction, immune depletion, and dysbiosis. Furthermore, ART interruption results in reservoir reactivation and rebound viremia. Both IL-21 and anti-α4β7 improve gut barrier functions, and we hypothesized that combining them would synergize as a dual therapy to improve immunological outcomes in SIV-infected rhesus macaques (RMs). We found no significant differences in CD4+ T cell reservoir size by intact proviral DNA assay. SIV rebounded in both dual-treated and control RMs following analytical therapy interruption (ATI), with time to rebound and initial rebound viremia comparable between groups; however, dual-treated RMs showed slightly better control of viral replication at the latest time points after ATI. Additionally, following ATI, dual-treated RMs showed immunological benefits, including T cell preservation and lower PD-1+ central memory T cell (TCM) frequency. Notably, PD-1+ TCMs were associated with reservoir size, which predicted viral loads (VLs) after ATI. Finally, 16S rRNA-Seq revealed better recovery from dysbiosis in treated animals, and the butyrate-producing Firmicute Roseburia predicted PD-1-expressing TCMs and VLs after ATI. PD-1+ TCMs and gut dysbiosis represent mechanisms of HIV persistence and pathogenesis, respectively. Therefore, combining IL-21 and anti-α4β7 may be an effective therapeutic strategy to improve immunological outcomes for people with HIV.
Collapse
Affiliation(s)
- Samuel D. Johnson
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center (UNMC), Omaha, Nebraska, USA
| | - Maria Pino
- Division of Microbiology and Immunology, Emory National Primate Research Center (ENPRC), Emory University, Atlanta, Georgia, USA
| | - Arpan Acharya
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center (UNMC), Omaha, Nebraska, USA
| | - Julien A. Clain
- Division of Microbiology and Immunology, Emory National Primate Research Center (ENPRC), Emory University, Atlanta, Georgia, USA
| | - Deepanwita Bose
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, Louisiana, USA
| | - Kevin Nguyen
- Division of Microbiology and Immunology, Emory National Primate Research Center (ENPRC), Emory University, Atlanta, Georgia, USA
| | - Justin Harper
- Division of Microbiology and Immunology, Emory National Primate Research Center (ENPRC), Emory University, Atlanta, Georgia, USA
| | - Francois Villinger
- New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, Louisiana, USA
| | - Mirko Paiardini
- Division of Microbiology and Immunology, Emory National Primate Research Center (ENPRC), Emory University, Atlanta, Georgia, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Siddappa N. Byrareddy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center (UNMC), Omaha, Nebraska, USA
- Department of Genetics, Cell Biology and Anatomy, and
- Department of Biochemistry and Molecular Biology, UNMC, Omaha, Nebraska, USA
| |
Collapse
|
28
|
Shin JJ, Shin HJ, Gutierrez A, Yoo N, Par-Young J, Osmani L, Shin MS, Sanchez-Lara PA, Bucala R, Soffer G, Kang I. Decreased T helper 1 cell function underlies recurrent sinopulmonary infections in the 17q12 deletion syndrome. EBioMedicine 2025; 112:105578. [PMID: 39891996 PMCID: PMC11840234 DOI: 10.1016/j.ebiom.2025.105578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 01/15/2025] [Accepted: 01/17/2025] [Indexed: 02/03/2025] Open
Abstract
BACKGROUND The 17q12 deletion syndrome (17q12DS) is a heterozygous deletion of a 1.4 megabase‒spanning DNA sequence on chromosome 17. The clinical characteristics of 17q12DS include neurodevelopmental disorders, kidney and urinary tract abnormalities. In our cohort of 37 subjects with 17q12DS, we observed increased atopic disorders and recurrent sinopulmonary infections, raising the possibility of immune dysregulation in 17q12DS, a feature that has not been previously reported. We tested the hypothesis that individuals with 17q12DS have altered T-cell function, contributing to recurrent infections and atopy. METHODS Alteration of CD4+ T-cell functions was assessed on FACS sorted CD4+ T-cells using RNA-seq analysis, and flow cytometry and multiplex assays. FINDINGS We found that individuals with 17q12DS had a substantially decreased frequency of CD4+ T-cells producing the T helper (Th) 1 cytokine IFN-γ but not Th2 and Th17 cytokines when compared to age-matched healthy controls (HCs). RNA-seq analysis of CD4+ T-cells from subjects with 17q12DS, when compared to HCs, revealed decreased levels of TBX21 encoding the Th1 transcription factor T-bet, IFNG, and other Th1 chemokine genes. These findings were validated using flow cytometry and multiplex assay. INTERPRETATION Our study is the first to demonstrate immune alterations in 17q12DS characterized by decreased T-bet and its downstream effector cytokines such as IFN-γ. These findings warrant further investigation into underlying mechanisms, which would inform precision therapy for individuals with 17q12DS. FUNDING National Institutes of HealthKL2 TR001862 to JJS, T35DK104689 to AG, 5T32AR007107 to JPY and LO, 1R01AG056728 to IK, and 1R21AI161838 to IK and JJS.
Collapse
Affiliation(s)
- Junghee J Shin
- Section of Rheumatology, Allergy & Immunology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA; Section of Pulmonary, Allergy, Immunology and Sleep Medicine, Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06520, USA.
| | - Hyeon Jun Shin
- Section of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Alan Gutierrez
- Section of Rheumatology, Allergy & Immunology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Noelle Yoo
- Section of Rheumatology, Allergy & Immunology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Jennefer Par-Young
- Section of Rheumatology, Allergy & Immunology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Lais Osmani
- Section of Rheumatology, Allergy & Immunology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Min Sun Shin
- Section of Rheumatology, Allergy & Immunology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Pedro A Sanchez-Lara
- Section of Medical Genetics, Department of Pediatrics, Guerin Children's at Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Richard Bucala
- Section of Rheumatology, Allergy & Immunology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Gary Soffer
- Section of Pulmonary, Allergy, Immunology and Sleep Medicine, Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Insoo Kang
- Section of Rheumatology, Allergy & Immunology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
29
|
Lu Y, Man XY. Diversity and function of regulatory T cells in health and autoimmune diseases. J Autoimmun 2025; 151:103357. [PMID: 39805189 DOI: 10.1016/j.jaut.2025.103357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 12/31/2024] [Accepted: 01/04/2025] [Indexed: 01/16/2025]
Abstract
Regulatory T cell (Treg) play a pivotal role in immune regulation and maintaining host immune homeostasis. Treg heterogeneity, characterized by diverse gene expression profiles and functional states, is complex in both health and disease. Research reveals that Tregs are not a uniform population but exhibit diversity based on their origin, location, and functional status. This heterogeneity is crucial for understanding Treg roles in various pathological conditions. Dysfunctional Tregs are closely linked to the pathogenesis of autoimmune diseases, although the precise mechanisms remain unclear. The phenotypic and functional heterogeneity of Tregs is particularly significant in diseases such as systemic lupus erythematosus, multiple sclerosis, rheumatoid arthritis, inflammatory bowel disease, type 1 diabetes, psoriasis and autoimmune liver diseases. This review explores Treg origins, classifications, and heterogeneity in these conditions, aiming to provide new perspectives and strategies for diagnosis and treatment. Understanding Treg heterogeneity and plasticity promises to reveal novel therapeutic targets and advance precision immunotherapy development.
Collapse
Affiliation(s)
- Yi Lu
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China
| | - Xiao-Yong Man
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, China.
| |
Collapse
|
30
|
Shin D, Gong J, Jeong SD, Cho Y, Kim H, Kim T, Cho K. Attractor Landscape Analysis Reveals a Reversion Switch in the Transition of Colorectal Tumorigenesis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412503. [PMID: 39840939 PMCID: PMC11848608 DOI: 10.1002/advs.202412503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/27/2024] [Indexed: 01/23/2025]
Abstract
A cell fate change such as tumorigenesis incurs critical transition. It remains a longstanding challenge whether the underlying mechanism can be unraveled and a molecular switch that can reverse such transition is found. Here a systems framework, REVERT, is presented with which can reconstruct the core molecular regulatory network model and a reversion switch based on single-cell transcriptome data over the transition process is identified. The usefulness of REVERT is demonstrated by applying it to single-cell transcriptome of patient-derived matched organoids of colon cancer and normal colon. REVERT is a generic framework that can be applied to investigate various cell fate transition phenomena.
Collapse
Affiliation(s)
- Dongkwan Shin
- Department of Bio and Brain EngineeringKorea Advanced Institute of Science and Technology (KAIST)Daejeon34141Republic of Korea
- Research InstituteNational Cancer CenterGoyang10408Republic of Korea
- Department of Cancer Biomedical ScienceNational Cancer Center Graduate School of Cancer Science and PolicyGoyang10408Republic of Korea
| | - Jeong‐Ryeol Gong
- Department of Bio and Brain EngineeringKorea Advanced Institute of Science and Technology (KAIST)Daejeon34141Republic of Korea
| | - Seoyoon D. Jeong
- Department of Bio and Brain EngineeringKorea Advanced Institute of Science and Technology (KAIST)Daejeon34141Republic of Korea
| | - Youngwon Cho
- Department of Molecular Medicine and Biopharmaceutical SciencesGraduate School of Convergence Science and TechnologySeoul National UniversitySeoul03080Republic of Korea
| | - Hwang‐Phill Kim
- Department of Molecular Medicine and Biopharmaceutical SciencesGraduate School of Convergence Science and TechnologySeoul National UniversitySeoul03080Republic of Korea
| | - Tae‐You Kim
- Department of Molecular Medicine and Biopharmaceutical SciencesGraduate School of Convergence Science and TechnologySeoul National UniversitySeoul03080Republic of Korea
| | - Kwang‐Hyun Cho
- Department of Bio and Brain EngineeringKorea Advanced Institute of Science and Technology (KAIST)Daejeon34141Republic of Korea
| |
Collapse
|
31
|
Xu Y, Wang Z, Li S, Su J, Gao L, Ou J, Lin Z, Luo OJ, Xiao C, Chen G. An in-depth understanding of the role and mechanisms of T cells in immune organ aging and age-related diseases. SCIENCE CHINA. LIFE SCIENCES 2025; 68:328-353. [PMID: 39231902 DOI: 10.1007/s11427-024-2695-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 07/28/2024] [Indexed: 09/06/2024]
Abstract
T cells play a critical and irreplaceable role in maintaining overall health. However, their functions undergo alterations as individuals age. It is of utmost importance to comprehend the specific characteristics of T-cell aging, as this knowledge is crucial for gaining deeper insights into the pathogenesis of aging-related diseases and developing effective therapeutic strategies. In this review, we have thoroughly examined the existing studies on the characteristics of immune organ aging. Furthermore, we elucidated the changes and potential mechanisms that occur in T cells during the aging process. Additionally, we have discussed the latest research advancements pertaining to T-cell aging-related diseases. These findings provide a fresh perspective for the study of T cells in the context of aging.
Collapse
Affiliation(s)
- Yudai Xu
- Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, 510632, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Zijian Wang
- Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, 510632, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Shumin Li
- Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, 510632, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Jun Su
- First Affiliated Hospital, Jinan University, Guangzhou, 510630, China
| | - Lijuan Gao
- Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, 510632, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, China
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Junwen Ou
- Anti Aging Medical Center, Clifford Hospital, Guangzhou, 511495, China
| | - Zhanyi Lin
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Oscar Junhong Luo
- Department of Systems Biomedical Sciences, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Chanchan Xiao
- Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, 510632, China.
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, China.
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou, 510632, China.
- The Sixth Affiliated Hospital of Jinan University (Dongguan Eastern Central Hospital), Jinan University, Dongguan, 523000, China.
- Zhuhai Institute of Jinan University, Jinan University, Zhuhai, 519070, China.
| | - Guobing Chen
- Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, 510632, China.
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, China.
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, School of Medicine, Jinan University, Guangzhou, 510632, China.
- The Sixth Affiliated Hospital of Jinan University (Dongguan Eastern Central Hospital), Jinan University, Dongguan, 523000, China.
- Zhuhai Institute of Jinan University, Jinan University, Zhuhai, 519070, China.
| |
Collapse
|
32
|
Satomi H, Kobayashi M, Ito I, Asano K, Makino M, Kawaguchi K, Kanno H. Predominance of T-bet-positive Th1 cells in infiltrating T-lymphocytes in most of active arteritis lesions of giant cell arteritis. Virchows Arch 2025; 486:267-275. [PMID: 38478104 DOI: 10.1007/s00428-024-03776-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: 08/04/2023] [Revised: 01/17/2024] [Accepted: 03/05/2024] [Indexed: 03/04/2025]
Abstract
Immunological mechanisms through the activation of CD4-positive T-cells have been assumed to be involved in the pathogenesis of giant cell arteritis (GCA). Many studies employing frozen tissues of temporal artery biopsy, peripheral blood lymphocytes, and plasma of GCA patients have revealed the contribution of interferon-γ and interleukin-17 in both protein and mRNA levels. However, the analyses using formalin-fixed and paraffin-embedded (FFPE) tissue specimens, in which the correlation between histopathologic pictures and immunological circumstances would be elucidated, have been limited. Here, we performed the immunohistochemical analyses of infiltrating small lymphocytes in GCA lesions using FFPE specimens, especially of the subsets of CD4-positive T-cells by immunohistochemistry with antibodies against T-bet, GATA-3, RORγT, and Foxp3, which is the differentiation-specific transcription factor for Th1, Th2, Th17, and Treg cells, respectively. In these slides, the nuclear-positive staining is much more clearly and easily identifiable than the cytoplasmic staining for cytokines. The results indicate the predominance of T-bet-positive Th1 cells in infiltrating T-cells in most of active arteritis lesions of GCA. Furthermore, our data suggest the possible immunosuppressive microenvironment induced by T-reg cells and M2-type macrophages in the arteritis lesions throughout the course of GCA inflammation.
Collapse
Affiliation(s)
- Hidetoshi Satomi
- Department of Pathology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
- Department of Pathology, Nagano Red Cross Hospital, 5-22-1 Wakasato, Nagano, 380-8582, Japan
- Department of Diagnostic Pathology and Cytology, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-Ku, Osaka, 541-8567, Japan
| | - Mikiko Kobayashi
- Department of Pathology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan
- Department of Pathology, Marunouchi Hospital, 1-7-45 Nagisa, Matsumoto, 390-8601, Japan
| | - Ichiro Ito
- Department of Pathology, Nagano Red Cross Hospital, 5-22-1 Wakasato, Nagano, 380-8582, Japan
| | - Koji Asano
- Department of Pathology, Suwa Central Hospital, 4300, Tamagawa, Chino, 391-8503, Japan
| | - Mutsuki Makino
- Department of Pathology, Shinonoi General Hospital, 666-1, Shinonoi, Nagano, 388-8004, Japan
| | - Kenji Kawaguchi
- Department of Pathology, Shinonoi General Hospital, 666-1, Shinonoi, Nagano, 388-8004, Japan
| | - Hiroyuki Kanno
- Department of Pathology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, 390-8621, Japan.
| |
Collapse
|
33
|
Cardani-Boulton A, Lin F, Bergmann CC. CD6 regulates CD4 T follicular helper cell differentiation and humoral immunity during murine coronavirus infection. J Virol 2025; 99:e0186424. [PMID: 39679790 PMCID: PMC11784103 DOI: 10.1128/jvi.01864-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 11/25/2024] [Indexed: 12/17/2024] Open
Abstract
During activation, the T cell transmembrane receptor CD6 becomes incorporated into the T cell immunological synapse where it can exert both co-stimulatory and co-inhibitory functions. Given the ability of CD6 to carry out opposing functions, this study sought to determine how CD6 regulates early T cell activation in response to viral infection. Infection of CD6-deficient mice with a neurotropic murine coronavirus resulted in greater activation and expansion of CD4 T cells in the draining lymph nodes. Further analysis demonstrated that there was also preferential differentiation of CD4 T cells into T follicular helper cells, resulting in accelerated germinal center responses and emergence of high-affinity virus-specific antibodies. Given that CD6 conversely supports CD4 T cell activation in many autoimmune models, we probed potential mechanisms of CD6-mediated suppression of CD4 T cell activation during viral infection. Analysis of CD6 binding proteins revealed that infection-induced upregulation of Ubash3a, a negative regulator of T cell receptor (TCR) signaling, was hindered in CD6-deficient lymph nodes. Consistent with greater T cell activation and reduced UBASH3a activity, the T cell receptor signal strength was intensified in CD6-deficient CD4 T cells. These results reveal a novel immunoregulatory role for CD6 in limiting CD4 T cell activation and deterring CD4 T follicular helper cell differentiation, thereby attenuating antiviral humoral immunity. IMPORTANCE CD6 monoclonal blocking antibodies are being therapeutically administered to inhibit T cell activation in autoimmune disorders. However, the multifaceted nature of CD6 allows for multiple and even opposing functions under different circumstances of T cell activation. We therefore sought to characterize how CD6 regulates T cell activation in the context of viral infections using an in vivo murine coronavirus model. In contrast to its role in autoimmunity, but consistent with its function in the presence of superantigens, we found that CD6 deficiency enhances CD4 T cell activation and CD4 T cell help to germinal center-dependent antiviral humoral responses. Finally, we provide evidence that CD6 regulates transcription of its intracellular binding partner UBASH3a, which suppresses T cell receptor (TCR) signaling and consequently T cell activation. These findings highlight the context-dependent flexibility of CD6 in regulating in vivo adaptive immune responses, which may be targeted to enhance antiviral immunity.
Collapse
MESH Headings
- Animals
- Mice
- Immunity, Humoral/immunology
- Cell Differentiation/immunology
- T Follicular Helper Cells/immunology
- Lymphocyte Activation/immunology
- Antigens, Differentiation, T-Lymphocyte/genetics
- Antigens, Differentiation, T-Lymphocyte/immunology
- Antigens, Differentiation, T-Lymphocyte/metabolism
- Coronavirus Infections/immunology
- Coronavirus Infections/virology
- Mice, Inbred C57BL
- Germinal Center/immunology
- Mice, Knockout
- CD4-Positive T-Lymphocytes/immunology
- Antigens, CD/immunology
- Antigens, CD/genetics
- T-Lymphocytes, Helper-Inducer/immunology
- Signal Transduction
- Murine hepatitis virus/immunology
- Antibodies, Viral/immunology
Collapse
Affiliation(s)
- Amber Cardani-Boulton
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Feng Lin
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Department of Immunity and Inflammation, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Cornelia C. Bergmann
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine of Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| |
Collapse
|
34
|
Wang CM, Chen YJ, Yang BC, Yang JW, Wang W, Zeng Y, Jiang J. Supplementation with active vitamin D3 ameliorates experimental autoimmune thyroiditis in mice by modulating the differentiation and functionality of intrathyroidal T-cell subsets. Front Immunol 2025; 16:1528707. [PMID: 39949783 PMCID: PMC11821646 DOI: 10.3389/fimmu.2025.1528707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 01/15/2025] [Indexed: 02/16/2025] Open
Abstract
Objective People with Hashimoto's thyroiditis (HT) often have low vitamin D3 concentrations. Some research has suggested that vitamin D3 supplementation reduces thyroid inflammation, but this remains controversial. Methods EAT was induced in female NOD/ShiLtJ mice by giving them water containing 0.05% sodium iodide, and 1μg/kg of 1α,25-(OH)2D3 was injected intraperitoneally every other day. After 8 weeks, the morphological architecture of the mouse thyroid follicles was examined by histological sections, thyroid autoantibodies and thyroid hormone concentrations were determined by enzyme-linked immunosorbent assays (ELISAs), and the major functions and subsets of B- and T-lymphocytes in the mouse thyroid were determined by tissue multiple immunofluorescence technology and ELISA. Results EAT caused thyroiditis follicle destruction and interfollicular lymphocyte infiltration in mice, increased concentrations of circulating thyroid autoimmune antibodies TG-Ab and TPO-Ab, and abnormal thyroid hormone levels. EAT also increased the number and functionality of CD4+ Tfh, Th17,Th1 and Th2 cells in the thyroid, while decreasing the number and functionality of CD4+ Treg cells and CD19+B10 cells. Treatment with VD3 reversed these changes. Conclusion Vitamin D3 supplementation can effectively treat autoimmune thyroiditis in mice. VD3 reduces autoimmune thyroid damage and decreases serum thyroid antibody levels in mice by inhibiting the differentiation and functionality of pro-inflammatory Tfh, Th17, Th1 and Th2 cells and by facilitating the differentiation and functionality of anti-inflammatory B10 cells and Treg.
Collapse
Affiliation(s)
- Chun-Mei Wang
- Department of General Surgery (Thyroid Surgery), Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Ying-Jie Chen
- Department of General Surgery (Thyroid Surgery), Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Bo-Cheng Yang
- Department of General Surgery (Thyroid Surgery), Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Jia-Wen Yang
- Department of General Surgery (Thyroid Surgery), Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Wei Wang
- Department of Thyroid, Head, Neck and Maxillofacial Surgery, Mianyang Third People’s Hospital, Mianyang, Sichuan, China
| | - Yang Zeng
- Department of Orthodontics, Affiliated Stomatology Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Jun Jiang
- Department of General Surgery (Thyroid Surgery), Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
35
|
Osum KC, Becker SH, Krueger PD, Mitchell JS, Hong SW, Magill IR, Jenkins MK. A minority of Th1 and Tfh effector cells express survival genes shared by memory cell progeny that require IL-7 or TCR signaling to persist. Cell Rep 2025; 44:115111. [PMID: 39723889 PMCID: PMC12009130 DOI: 10.1016/j.celrep.2024.115111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 10/24/2024] [Accepted: 12/03/2024] [Indexed: 12/28/2024] Open
Abstract
It is not clear how CD4+ memory T cells are formed from a much larger pool of earlier effector cells. We found that transient systemic bacterial infection rapidly generates several antigen-specific T helper (Th)1 and T follicular helper (Tfh) cell populations with different tissue residence behaviors. Although most cells of all varieties had transcriptomes indicative of cell stress and death at the peak of the response, some had already acquired a memory cell signature characterized by expression of genes involved in cell survival. Each Th1 and Tfh cell type was maintained long term by interleukin (IL)-7, except germinal center Tfh cells, which depended on a T cell antigen receptor (TCR) signal. The results indicate that acute infection induces rapid differentiation of Th1 and Tfh cells, a minority of which quickly adopt the gene expression profile of memory cells and survive by signals from the IL-7 receptor or TCR.
Collapse
Affiliation(s)
- Kevin C Osum
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Samuel H Becker
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Peter D Krueger
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Jason S Mitchell
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Sung-Wook Hong
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA; Department of Biotechnology, Yonsei University, Seoul, South Korea
| | - Ian R Magill
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Marc K Jenkins
- Department of Microbiology and Immunology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455, USA.
| |
Collapse
|
36
|
Lee JS, Lacerda E, Kingdon C, Susannini G, Dockrell HM, Nacul L, Cliff JM. Abnormal T-Cell Activation And Cytotoxic T-Cell Frequency Discriminates Symptom Severity In Myalgic Encephalomyelitis/Chronic Fatigue Syndrome. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.01.02.24319359. [PMID: 39830245 PMCID: PMC11741448 DOI: 10.1101/2025.01.02.24319359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is a debilitating but poorly-understood disease. ME/CFS symptoms can range from mild to severe, and include immune system effects alongside incapacitating fatigue and post-exertional disease exacerbation. In this study, we examined immunological profiles of people living with ME/CFS by flow cytometry, focusing on cytotoxic cells, to determine whether people with mild/moderate (n= 43) or severe ME/CFS (n=53) expressed different immunological markers. We found that people with mild/moderate ME/CFS had increased expression of cytotoxic effector molecules alongside enhanced proportions of early-immunosenescence cells, determined by the CD28 - CD57 - phenotype, indicative of persistent viral infection. In contrast, people with severe ME/CFS had higher proportions of activated circulating lymphocytes, determined by CD69 + and CD38 + expression, and expressed more pro-inflammatory cytokines, including IFNγ, TNF and IL-17, following stimulation in vitro , indicative of prolonged non-specific inflammation. These changes were consistent across different cell types including CD8 + T cells, mucosal associated invariant T cells and Natural Killer cells, indicating generalised altered cytotoxic responses across the innate and adaptive immune system. These immunological differences likely reflect different disease pathogenesis mechanisms occurring in the two clinical groups, opening up opportunities for the development of prognostic markers and stratified treatments.
Collapse
|
37
|
Yao X, Huo W, Wang Y, Xia D, Chen Y, Tang Y, Tang H, Yang W, Liu Y, Xue J, Yuan Q, Gao X, Cao K. Environmental Low-Dose Radiation Activates Th1 Immunity through the Mitochondria-STING Pathway. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:22907-22918. [PMID: 39689952 DOI: 10.1021/acs.est.4c08009] [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: 12/19/2024]
Abstract
The presence of low-dose radiation (LDR) in the environment has become more prevalent. However, the effect of LDR exposure on the immune system remains elusive. Here, we interestingly found that LDR specifically elevated the percentage of CD4+IFNγ+ Th1 splenocytes, both in vitro and in vivo, without affecting the percentage of CD8+IFNγ+ Tc1 cells and regulatory T cells. A similar phenomenon was found in T cells from peripheral blood. Mechanistically, we found that LDR can induce mitochondrial damage, which stimulated the STING signaling pathway, leading to the enhanced expression of T-bet, the master transcriptional factor of Th1-cell differentiation. The specific STING signal inhibitor can abrogate the effect of LDR on Th1 differentiation, confirming the central role of the STING pathway. To further validate the immunoregulatory role of LDR, we exposed mice with whole body LDR and evaluated if LDR could protect mice against triple-negative breast cancer through enhanced antitumor immunity. As expected, LDR significantly delayed tumor development and promoted cell death. Meanwhile, LDR resulted in increased tumor-infiltrating Th1 cells, while the proportion of Tc1 and Treg cells remained unchanged. Furthermore, the infiltration of antitumor macrophages was also increased. In summary, we revealed that environmental LDR could specifically regulate Th1 T-cell activities, providing critical information for the potential application of LDR in both clinical and nonclinical settings.
Collapse
Affiliation(s)
- Xiuxiu Yao
- College of Materials Science and Engineering, Beijing University of Technology, Beijing 100124, China
| | - Wendi Huo
- College of Materials Science and Engineering, Beijing University of Technology, Beijing 100124, China
| | - Yuchen Wang
- Department of Chemistry, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Dongfang Xia
- College of Materials Science and Engineering, Beijing University of Technology, Beijing 100124, China
| | - Yan Chen
- College of Materials Science and Engineering, Beijing University of Technology, Beijing 100124, China
| | - Yuhua Tang
- College of Materials Science and Engineering, Beijing University of Technology, Beijing 100124, China
| | - Huayong Tang
- Department of Chemistry, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Wenjiang Yang
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
| | - Yu Liu
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
| | - Jingquan Xue
- Beijing Engineering Research Center of Radiographic Techniques and Equipment, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
| | - Qing Yuan
- Department of Chemistry, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Xueyun Gao
- Department of Chemistry, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| | - Kai Cao
- Department of Chemistry, College of Chemistry and Life Science, Beijing University of Technology, Beijing 100124, China
| |
Collapse
|
38
|
Shinzawa Y, Hara D, Shinguryo Y, Yokoyama S, Kawada M, Hayakawa Y. PP2A negatively regulates NK cell T-bet expression and anti-tumor effector function. Int Immunol 2024; 37:97-107. [PMID: 39404747 DOI: 10.1093/intimm/dxae057] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 10/11/2024] [Indexed: 12/28/2024] Open
Abstract
The transcription factor T-bet is essential for the anti-tumor effector function of natural killer (NK) cells, but the mechanism regulating its expression in NK cells remains unclear. In this study, we aimed to identify an NK cell-intrinsic regulator that controls T-bet expression. Using T-bet-luciferase reporter assay screening, we identified a protein phosphatase inhibitor as a potential activator of T-bet expression. A series of protein phosphatase 2A (PP2A)-specific inhibitors (PP2Ai) or PP2A siRNA induced the expression of T-bet. In PP2Ai-treated mice, the expression of T-bet and its downstream effector molecules, granzyme B and IFN-γ, was also upregulated in NK cells. Mechanistically, PP2Ai increased the phosphorylation of mTOR and ribosomal protein S6 in NK cells, and mTOR inhibitor canceled the effects of PP2Ai in NK cells. Importantly, NK cells isolated from PP2Ai-treated mice showed higher cytotoxicity and IFN-γ production; therefore, they increased the anti-tumor effector function of NK cells. Accordingly, PP2Ai treatment inhibited lung metastasis of B16 melanoma by NK cell- and mTOR-dependent mechanisms. These results suggest that PP2A negatively regulates NK cell T-bet expression and effector function by an mTOR-dependent mechanism.
Collapse
Affiliation(s)
- Yui Shinzawa
- Section of Host Defences, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | - Daisuke Hara
- Section of Host Defences, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | - Yuki Shinguryo
- Section of Host Defences, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | - Satoru Yokoyama
- Department of Cancer Cell Biology, Faculty of Pharmaceutical Sciences, University of Toyama, Toyama, Japan
| | - Manabu Kawada
- Laboratory of Oncology, Institute of Microbial Chemistry, Tokyo, Japan
| | - Yoshihiro Hayakawa
- Section of Host Defences, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| |
Collapse
|
39
|
Liao Y, Zheng Y, Zhang R, Chen X, Huang J, Liu J, Zhao Y, Zheng Y, Zhang X, Gao Z, Gao X, Bu J, Peng T, Li X, Shen E. Regulatory roles of transcription factors T-bet and Eomes in group 1 ILCs. Int Immunopharmacol 2024; 143:113229. [PMID: 39357208 DOI: 10.1016/j.intimp.2024.113229] [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: 07/06/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 10/04/2024]
Abstract
T-bet and Eomes, both T-box transcription factors, have been extensively studied for their critical roles in the differentiation and functional maintenance of various immune cells. In this review, we provide a focused overview of their contributions to the transcriptional activation and differentiation, development, and terminal maturation of natural killer cells and innate lymphoid cell 1 cells. Furthermore, the interplay between T-bet and Eomes in regulating NK cell function, and its subsequent implications for immune responses against infections and tumors, is thoroughly examined. The review explores the ramifications of dysregulated transcription factor expression, examining its impact on homeostatic balance and its role in a spectrum of disease models. Expression variances among distinct NK cell subsets resident in different tissues are highlighted to underscore the complexity of their biological roles. Collectively, this work aims to expand the current understanding of NK cell biology, thereby paving the way for innovative approaches in the realm of NK cell-based immunotherapies.
Collapse
Affiliation(s)
- Yue Liao
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China
| | - Yanling Zheng
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China; Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ruizhi Zhang
- Department of Emergency Medicine, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiangming Chen
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China
| | - Jijun Huang
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China
| | - Jiamin Liu
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China
| | - Yuyang Zhao
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China
| | - Yu Zheng
- National Center for STD Control, Chinese Center for Disease Control and Prevention, Nanjing, Jiangsu, China; Hospital for Skin Disease (Institute of Dermatology), Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China
| | - Xueyan Zhang
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China
| | - Zhiyan Gao
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China
| | - Xiaojing Gao
- National Center for STD Control, Chinese Center for Disease Control and Prevention, Nanjing, Jiangsu, China; Hospital for Skin Disease (Institute of Dermatology), Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China
| | - Jin Bu
- National Center for STD Control, Chinese Center for Disease Control and Prevention, Nanjing, Jiangsu, China; Hospital for Skin Disease (Institute of Dermatology), Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China.
| | - Tieli Peng
- The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, China.
| | - Xiaomin Li
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China.
| | - Erxia Shen
- Sino-French Hoffmann Institute, School of Basic Medical Sciences, The Second Affiliated Hospital of Guangzhou Medical University, State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
40
|
Hao ZN, Tan XP, Zhang Q, Li J, Xia R, Ma Z. Lactate and Lactylation: Dual Regulators of T-Cell-Mediated Tumor Immunity and Immunotherapy. Biomolecules 2024; 14:1646. [PMID: 39766353 PMCID: PMC11674224 DOI: 10.3390/biom14121646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/14/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
Lactate and its derivative, lactylation, play pivotal roles in modulating immune responses within the tumor microenvironment (TME), particularly in T-cell-mediated cancer immunotherapy. Elevated lactate levels, a hallmark of the Warburg effect, contribute to immune suppression through CD8+ T cell functionality and by promoting regulatory T cell (Treg) activity. Lactylation, a post-translational modification (PTM), alters histone and non-histone proteins, influencing gene expression and further reinforcing immune suppression. In the complex TME, lactate and its derivative, lactylation, are not only associated with immune suppression but can also, under certain conditions, exert immunostimulatory effects that enhance cytotoxic responses. This review describes the dual roles of lactate and lactylation in T-cell-mediated tumor immunity, analyzing how these factors contribute to immune evasion, therapeutic resistance, and immune activation. Furthermore, the article highlights emerging therapeutic strategies aimed at inhibiting lactate production or disrupting lactylation pathways to achieve a balanced regulation of these dual effects. These strategies offer new insights into overcoming tumor-induced immune suppression and hold the potential to improve the efficacy of cancer immunotherapies.
Collapse
Affiliation(s)
- Zhi-Nan Hao
- Department of Gastroenterology, First Affiliated Hospital of Yangtze University, Health Science Center, Yangtze University, Jingzhou 434023, China; (Z.-N.H.); (Q.Z.); (J.L.)
- Digestive Disease Research Institution of Yangtze University, Yangtze University, Jingzhou 434023, China;
| | - Xiao-Ping Tan
- Digestive Disease Research Institution of Yangtze University, Yangtze University, Jingzhou 434023, China;
- The Third Clinical Medical College of Yangtze University, Jingzhou Hospital of Traditional Chinese Medicine, Jingzhou 434023, China
| | - Qing Zhang
- Department of Gastroenterology, First Affiliated Hospital of Yangtze University, Health Science Center, Yangtze University, Jingzhou 434023, China; (Z.-N.H.); (Q.Z.); (J.L.)
- Digestive Disease Research Institution of Yangtze University, Yangtze University, Jingzhou 434023, China;
| | - Jie Li
- Department of Gastroenterology, First Affiliated Hospital of Yangtze University, Health Science Center, Yangtze University, Jingzhou 434023, China; (Z.-N.H.); (Q.Z.); (J.L.)
- Digestive Disease Research Institution of Yangtze University, Yangtze University, Jingzhou 434023, China;
| | - Ruohan Xia
- Department of Gastroenterology, First Affiliated Hospital of Yangtze University, Health Science Center, Yangtze University, Jingzhou 434023, China; (Z.-N.H.); (Q.Z.); (J.L.)
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China
| | - Zhaowu Ma
- Department of Gastroenterology, First Affiliated Hospital of Yangtze University, Health Science Center, Yangtze University, Jingzhou 434023, China; (Z.-N.H.); (Q.Z.); (J.L.)
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China
| |
Collapse
|
41
|
Qin Y, Qian Y, Liu S, Chen R. A double-edged sword role of IFN-γ-producing iNKT cells in sepsis: Persistent suppression of Treg cell formation in an Nr4a1-dependent manner. iScience 2024; 27:111462. [PMID: 39720538 PMCID: PMC11667017 DOI: 10.1016/j.isci.2024.111462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 10/17/2024] [Accepted: 11/20/2024] [Indexed: 12/26/2024] Open
Abstract
Sepsis, a leading cause of mortality in intensive care units worldwide, lacks effective treatments for advanced-stage sepsis. Therefore, understanding the underlying mechanisms of this disease is crucial. This study reveals that invariant natural killer T (iNKT) cells have an opposing role in the progression of sepsis by suppressing regulatory T (Treg) cell differentiation and function. The activation of iNKT cells by α-Galcer enhances interferon (IFN)-γ production. Blocking antibodies or transferring IFN-γ-deficient iNKT cells demonstrates that iNKT cells inhibit Treg differentiation through IFN-γ production. Additionally, iNKT cell-mediated Treg inhibition prevents secondary infection caused by Listeria monocytogenes during the post-septic phase. The transcriptomic analysis of Treg cells further reveals that the suppressive function of Tregs is impaired by iNKT cells. Finally, we demonstrate that iNKT cells inhibit Treg differentiation in an Nr4a1-dependent manner. Our data uncover the dual function of iNKT cells in sepsis progression and provide a potential treatment target for this adverse long-term outcome induced by sepsis.
Collapse
Affiliation(s)
- Yingyu Qin
- Department of Pathogenic Biology and Immunology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Yilin Qian
- Department of Pathogenic Biology and Immunology, Jiangsu Provincial Key Laboratory of Critical Care Medicine, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Shengqiu Liu
- Department of Pathogenic Biology and Immunology, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Rong Chen
- The Affiliated Zhongda Hospital, Clinical Medical College, Southeast University, Nanjing, Jiangsu, China
| |
Collapse
|
42
|
Xu J, Wang H, Zhang C, Jin SH, Chen X, Tan F, Frey B, Hecht M, Sun JG, Gaipl US, Ma H, Zhou JG. Efficacy of radiotherapy combined with atezolizumab or docetaxel in patients with previously treated NSCLC. iScience 2024; 27:111363. [PMID: 39640586 PMCID: PMC11617966 DOI: 10.1016/j.isci.2024.111363] [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: 08/20/2024] [Revised: 10/01/2024] [Accepted: 10/15/2024] [Indexed: 12/07/2024] Open
Abstract
Radiotherapy showed synergy with immunotherapy, yet the comparative effectiveness of combining immunotherapy (iRT) or chemotherapy (CRT) after platinum therapy failure in advanced non-small cell lung cancer (NSCLC) remains unexplored. We analyzed 163 patients (iRT: n = 120 vs. CRT: n = 43) eligible for combination radiotherapy. Before matching, median overall survival (OS) was significantly longer in iRT group (7.79 vs. 4.57 months, hazard ratio [HR]: 0.62, 95% confidence interval [CI]: 0.41-0.94, p = 0.024). After 1:2 propensity score matching (PSM) and inverse probability of treatment weighting (IPTW), iRT group showed improved OS, consistent with unmatched analysis (PSM, p = 0.033 and IPTW, p = 0.035). Exploratory analysis suggested that PD1+, central memory PD1+, and effector memory PD-L1+ CD4+ T cells were strong predictive biomarkers for iRT-treated patients (P OS = 0.025, P OS = 0.002, P OS = 0.010, respectively). Proliferative CD4+ T celllow was a prognostic (P OS = 0.008) and predictive biomarker for iRT (P OS < 0.001). Our work revealed iRT was prolonged OS in previously treated advanced NSCLC patients. Additionally, proliferative CD4+ T cell served as prognostic and predictive biomarkers.
Collapse
Affiliation(s)
- Junzhu Xu
- Department of Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi 563000, P.R. China
| | - Haitao Wang
- Thoracic Surgery Branch, Center for Cancer Research, NCI, NIH, Bethesda, MD 20892, USA
| | - Chi Zhang
- Department of Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi 563000, P.R. China
| | - Su-Han Jin
- Department of Orthodontics, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China
| | - Xiaofei Chen
- Oncology Biometrics, AstraZeneca, Gaithersburg, MD 20850, USA
| | - Fangya Tan
- Harrisburg University of Science and Technology, Harrisburg, PA 17101, USA
| | - Benjamin Frey
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
- FAU Profile Center Immunomedicine (FAU I-MED), Friedrich-Alexander- Universität Erlangen-Nurnberg, Erlangen, Germany
| | - Markus Hecht
- Department of Radiotherapy and Radiation Oncology, Saarland University Medical Center, Homburg, Germany
| | - Jian-Guo Sun
- Cancer Institute, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Udo S. Gaipl
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
- FAU Profile Center Immunomedicine (FAU I-MED), Friedrich-Alexander- Universität Erlangen-Nurnberg, Erlangen, Germany
| | - Hu Ma
- Department of Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi 563000, P.R. China
| | - Jian-Guo Zhou
- Department of Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi 563000, P.R. China
- Translational Radiobiology, Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN, Erlangen, Germany
- FAU Profile Center Immunomedicine (FAU I-MED), Friedrich-Alexander- Universität Erlangen-Nurnberg, Erlangen, Germany
| |
Collapse
|
43
|
Liu F, Bai Y, Wu X, Wan Y, Luo S, Zhang L, Li T, Tang H, Tang X, Chen R, Chen Q, Xie Y, Guo P. Network pharmacology combined with experimental validation reveals the mechanism of action of cangerzisan on allergic rhinitis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 335:118611. [PMID: 39053712 DOI: 10.1016/j.jep.2024.118611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/29/2024] [Accepted: 07/21/2024] [Indexed: 07/27/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Allergic rhinitis (AR) stands as a non-infectious inflammatory condition affecting the nasal mucosa, marked by bouts of sneezing, nasal itching, and congestion. This ailment afflicts individuals across all age groups and poses challenges for effective treatment due to its chronic nature. Cangerzisan (CEZS), documented in the Jishengfang compendium, represents a traditional Chinese medicinal formula long utilized for AR management. AIM OF THE STUDY Investigating mechanism beneath therapeutic effect of CEZS in alleviating AR. MATERIALS AND METHODS The main active components in CEZS were determined by High Performance Liquid Chromatography (HPLC).The active constituents of CEZS and their corresponding targets were identified through an exhaustive screening process employing TCMSP database. To identify targets relevant to AR, GeneCards, OMIM, and DisGeNET databases were thoroughly applied. Protein-protein interaction (PPI) network was assembled utilizing STRING platform. Potential signaling pathways influenced by CEZS were delineated through GO and KEGG enrichment analyses. Subsequently, an AR model was induced by administering aluminum hydroxide (Al(OH)3) and ovalbumin (OVA) for affecting basal and local sensitization, respectively, facilitating experimental validation of the principal signaling pathways. RESULTS There were 61 active constituents identified within CEZS, targeting a pool of 129 entities associated with AR treatment. Pathways analysis of KEGG revealed that CEZS potentially inhibits AR advancement via modulating TLR4 signaling pathway. Animal experiments demonstrated that CEZS effectively alleviated symptom scores in guinea pigs with AR. Moreover, it exhibited notable improvements in serum immune and inflammatory factors levels, as well as reduced inflammatory infiltration within nasal mucosa, including goblet and mast cells. CEZS was found to enhance GATA-3 expression while reducing T-bet expression, thereby modulating the TH1/TH2 immune balance. Additionally, CEZS downregulated HMGB1, TLR4, and p-NF-κB/NF-κB protein expressions within nasal mucosa of guinea pigs. CONCLUSIONS The therapeutic mechanism of CEZS against AR involves rectifying TH1/TH2 immune imbalance and upregulating inflammatory and immune factors through modulating key proteins expression within TLR4 pathway. This targeted regulation effectively impedes AR progression.
Collapse
Affiliation(s)
- Feifan Liu
- College of Ethnic Medicine, Yunnan University of Chinese Medicine, Kunming, 650500, Yunnan, China
| | - Yuanmei Bai
- College of Ethnic Medicine, Yunnan University of Chinese Medicine, Kunming, 650500, Yunnan, China
| | - Xue Wu
- College of Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, 650500, Yunnan, China
| | - Yan Wan
- College of Ethnic Medicine, Yunnan University of Chinese Medicine, Kunming, 650500, Yunnan, China
| | - Shifang Luo
- College of Ethnic Medicine, Yunnan University of Chinese Medicine, Kunming, 650500, Yunnan, China
| | - Linao Zhang
- College of Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, 650500, Yunnan, China
| | - Tao Li
- College of Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, 650500, Yunnan, China
| | - Hua Tang
- College of Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, 650500, Yunnan, China
| | - Xianjin Tang
- College of Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, 650500, Yunnan, China
| | - Rong Chen
- Yunnan Key Laboratory of Dai and Yi Medicines, Yunnan, Kunming, 650500, China
| | - Qinghua Chen
- College of Ethnic Medicine, Yunnan University of Chinese Medicine, Kunming, 650500, Yunnan, China.
| | - Yuhuan Xie
- College of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, 650500, Yunnan, China.
| | - Peixin Guo
- College of Ethnic Medicine, Yunnan University of Chinese Medicine, Kunming, 650500, Yunnan, China.
| |
Collapse
|
44
|
Sun S, Zhang Y, Gu X, Li D, Liu W, Zhang P, Niu J, Lu J, Chai L. Chinese herbal formula, modified Guilu Erxian glue, alleviates apoptosis of hematopoietic stem cells by regulating SLAM-SAP signal pathway in aplastic anemia mice model. JOURNAL OF ETHNOPHARMACOLOGY 2024; 335:118575. [PMID: 39009326 DOI: 10.1016/j.jep.2024.118575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 06/26/2024] [Accepted: 07/10/2024] [Indexed: 07/17/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Guilu Erxian Glue (GEG) and Danggui Buxue Tang (DBT) are traditional Chinese herbal formulas. According to the theory of traditional Chinese medicine, the combination of those two formulas (Modified Guilu Erxian Glue, MGEG) has the effects of tonifying the kidney and producing blood, was usually used to treat bone marrow failure diseases, including aplastic anemia (AA). AIM OF THE STUDY T lymphocytes play a crucial role in the disease pathogenesis and progression of AA. Our preliminary results confirmed that GEG can improve the damage of hematopoietic stem cells in mice, while DBT can reduce the proliferation and differentiation of T lymphocytes and inhibit the production of IFN-γ. We hypothesized that the combination of those two herbal formulas could inhibit immune attack and restore hematopoietic function through multiple mechanisms. In this study, we aim to study the curative effect of MGEG on regulating the expression of Signal lymphocyte activating molecule (SLAM), an activation-related molecule in T lymphocytes, thereby suppressing the immune function of T cells and decelerating the damage to hematopoietic stem cells. MATERIALS AND METHODS High-performance liquid chromatography-electrospray ionization/mass spectrometry system was used to identify the components of the MGEG formulation. Induction of aplastic anemia mouse model by injecting allogeneic lymphocyte suspension into BABL/c mice after ionizing radiation. Cyclosporine A (CsA) was used as a positive control drug. Flow cytometry was used to detect the number and apoptosis rate of hematopoietic stem cells in the bone marrow. Enzyme-linked immunosorbent assay was performed to measure the levels of IFN-γ and TNF-α. Immunofluorescence staining was used to assess the expression of T-bet and SLAM-SAP. Western Blot was conducted to examine the expression of activation-related molecules in T lymphocytes and proteins related to the Fas signal pathway. Hematoxylin-eosin staining was performed to observe pathological changes in the bone marrow tissue. Wright-Giemsa staining was utilized to evaluate alterations in the cellular composition and basic structure of the bone marrow cells (BMCs). Transmission electron microscopy was employed to observe changes in the structure and morphology of hematopoietic stem cells. The hematology analyzer was used to detect peripheral blood parameters. RESULTS Twenty-three different components were identified in MGEG. After MGEG treatment, the expression levels of Fyn and SLAM-SAP binding were increased in AA mice, while the expression levels of T-bet were decreased and the secretion of IFN-γ was reduced significantly. Additionally, MGEG also could downregulate the protein levels of Fas, caspase-3, and cleaved caspase-3 in AA mice. CONCLUSION MGEG could attenuate the production of IFN-γ by promoting the SLAM-SAP signal pathway to regulate the generation and distribution of T-bet in T cells. Additionally, it suppresses apoptosis of HSCs through intervention in the Fas-dependent pathway, thereby mitigating immune-mediated damage to HSCs.
Collapse
Affiliation(s)
- Song Sun
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Yingkai Zhang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Xin Gu
- Department of Pharmacy, Dongzhimen Hospital, Beijing University of Traditional Chinese Medicine, Beijing, 100700, China
| | - Dongyang Li
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Wei Liu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Pingxin Zhang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Jingmin Niu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Jinsong Lu
- Department of Pharmacy, Dongzhimen Hospital, Beijing University of Traditional Chinese Medicine, Beijing, 100700, China.
| | - Limin Chai
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China.
| |
Collapse
|
45
|
Akula S, Alvarado-Vazquez A, Haide Mendez Enriquez E, Bal G, Franke K, Wernersson S, Hallgren J, Pejler G, Babina M, Hellman L. Characterization of Freshly Isolated Human Peripheral Blood B Cells, Monocytes, CD4+ and CD8+ T Cells, and Skin Mast Cells by Quantitative Transcriptomics. Int J Mol Sci 2024; 25:13050. [PMID: 39684762 DOI: 10.3390/ijms252313050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 11/25/2024] [Accepted: 12/01/2024] [Indexed: 12/18/2024] Open
Abstract
Quantitative transcriptomics offers a new way to obtain a detailed picture of freshly isolated cells. By direct isolation, the cells are unaffected by in vitro culture, and the isolation at cold temperatures maintains the cells relatively unaltered in phenotype by avoiding activation through receptor cross-linking or plastic adherence. Simultaneous analysis of several cell types provides the opportunity to obtain detailed pictures of transcriptomic differences between them. Here, we present such an analysis focusing on four human blood cell populations and compare those to isolated human skin mast cells. Pure CD19+ peripheral blood B cells, CD14+ monocytes, and CD4+ and CD8+ T cells were obtained by fluorescence-activated cell sorting, and KIT+ human connective tissue mast cells (MCs) were purified by MACS sorting from healthy skin. Detailed information concerning expression levels of the different granule proteases, protease inhibitors, Fc receptors, other receptors, transcription factors, cell signaling components, cytoskeletal proteins, and many other protein families relevant to the functions of these cells were obtained and comprehensively discussed. The MC granule proteases were found exclusively in the MC samples, and the T-cell granzymes in the T cells, of which several were present in both CD4+ and CD8+ T cells. High levels of CD4 were also observed in MCs and monocytes. We found a large variation between the different cell populations in the expression of Fc receptors, as well as for lipid mediators, proteoglycan synthesis enzymes, cytokines, cytokine receptors, and transcription factors. This detailed quantitative comparative analysis of more than 780 proteins of importance for the function of these populations can now serve as a good reference material for research into how these entities shape the role of these cells in immunity and tissue homeostasis.
Collapse
Affiliation(s)
- Srinivas Akula
- Department of Cell and Molecular Biology, Uppsala University, The Biomedical Center, Box 596, SE-751 24 Uppsala, Sweden
- Department of Animal Biosciences, Swedish University of Agricultural Sciences, Box 7023, SE-75007 Uppsala, Sweden
| | - Abigail Alvarado-Vazquez
- Department of Medical Biochemistry and Microbiology, The Biomedical Center, Box 582, SE-75123 Uppsala, Sweden
| | - Erika Haide Mendez Enriquez
- Department of Medical Biochemistry and Microbiology, The Biomedical Center, Box 582, SE-75123 Uppsala, Sweden
| | - Gürkan Bal
- Institute of Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Kristin Franke
- Institute of Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Sara Wernersson
- Department of Animal Biosciences, Swedish University of Agricultural Sciences, Box 7023, SE-75007 Uppsala, Sweden
| | - Jenny Hallgren
- Department of Medical Biochemistry and Microbiology, The Biomedical Center, Box 582, SE-75123 Uppsala, Sweden
| | - Gunnar Pejler
- Department of Medical Biochemistry and Microbiology, The Biomedical Center, Box 582, SE-75123 Uppsala, Sweden
| | - Magda Babina
- Institute of Allergology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Immunology and Allergology IA, Hindenburgdamm 30, 12203 Berlin, Germany
| | - Lars Hellman
- Department of Cell and Molecular Biology, Uppsala University, The Biomedical Center, Box 596, SE-751 24 Uppsala, Sweden
| |
Collapse
|
46
|
Arroyo-Olarte RD, Flores-Castelán JC, Armas-López L, Escobedo G, Terrazas LI, Ávila-Moreno F, Leon-Cabrera S. Targeted Demethylation of FOXP3-TSDR Enhances the Suppressive Capacity of STAT6-deficient Inducible T Regulatory Cells. Inflammation 2024; 47:2159-2172. [PMID: 38700792 PMCID: PMC11606997 DOI: 10.1007/s10753-024-02031-4] [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: 01/28/2024] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 11/30/2024]
Abstract
In vitro induced T regulatory cells (iTregs) are promising for addressing inflammation-driven diseases. However, current protocols for the generation and expansion of iTregs fail to induce extensive demethylation of the Treg-specific demethylated region (TSDR) within the FOXP3 gene, recognized as the master regulator for regulatory T cells (Tregs). This deficiency results in the rapid loss of Foxp3 expression and an unstable regulatory phenotype. Nevertheless, inhibition of STAT6 signaling effectively stabilizes Foxp3 expression in iTregs. Thus, this study aimed to develop a protocol combining epigenetic editing with STAT6 deficiency to improve iTregs' ability to maintain stable suppressive function and a functional phenotype. Our findings demonstrate that the combination of STAT6 deficiency (STAT6-/-) with targeted demethylation of the TSDR using a CRISPR-TET1 tool leads to extensive demethylation of FOXP3-TSDR. Demethylation in STAT6-/- iTregs was associated with enhanced expression of Foxp3 and suppressive markers such as CTLA-4, PD-1, IL-10, and TGF-β. Furthermore, the edited STAT6-/- iTregs exhibited an increased capacity to suppress CD8+ and CD4+ lymphocytes and could more efficiently impair Th1-signature gene expression compared to conventional iTregs. In conclusion, the deactivation of STAT6 and TSDR-targeted demethylation via CRISPR-TET1 is sufficient to induce iTregs with heightened stability and increased suppressive capacity, offering potential applications against inflammatory and autoimmune diseases.
Collapse
Affiliation(s)
- Rubén D Arroyo-Olarte
- Unidad de Biomedicina. Facultad de Estudios Superiores-Iztacala, Universidad Nacional Autónoma de México, Av. De los Barrios 1, Los Reyes Iztacala, Edo. De México, Tlalnepantla, México
- Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Av. De los Barrios 1, Los Reyes Iztacala, Edo. De México, Tlalnepantla, México
| | - Juan C Flores-Castelán
- Unidad de Biomedicina. Facultad de Estudios Superiores-Iztacala, Universidad Nacional Autónoma de México, Av. De los Barrios 1, Los Reyes Iztacala, Edo. De México, Tlalnepantla, México
| | - Leonel Armas-López
- Unidad de Biomedicina. Facultad de Estudios Superiores-Iztacala, Universidad Nacional Autónoma de México, Av. De los Barrios 1, Los Reyes Iztacala, Edo. De México, Tlalnepantla, México
| | - Galileo Escobedo
- Laboratory of Immunometabolism, Research Division, General Hospital of Mexico "Dr. Eduardo Liceaga", 06720, Mexico City, Mexico
| | - Luis I Terrazas
- Unidad de Biomedicina. Facultad de Estudios Superiores-Iztacala, Universidad Nacional Autónoma de México, Av. De los Barrios 1, Los Reyes Iztacala, Edo. De México, Tlalnepantla, México
- Laboratorio Nacional en Salud, Facultad de Estudios Superiores-Iztacala, Universidad Nacional Autónoma de México, Edo. De México, Tlalnepantla, México
| | - Federico Ávila-Moreno
- Unidad de Biomedicina. Facultad de Estudios Superiores-Iztacala, Universidad Nacional Autónoma de México, Av. De los Barrios 1, Los Reyes Iztacala, Edo. De México, Tlalnepantla, México
| | - Sonia Leon-Cabrera
- Unidad de Biomedicina. Facultad de Estudios Superiores-Iztacala, Universidad Nacional Autónoma de México, Av. De los Barrios 1, Los Reyes Iztacala, Edo. De México, Tlalnepantla, México.
- Carrera de Médico Cirujano, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Av. De los Barrios 1, Los Reyes Iztacala, Edo. De México, Tlalnepantla, México.
| |
Collapse
|
47
|
Tebibi K, Ben Laamari R, Saied Z, Maghrebi O, Touzi H, Meddeb Z, Ben Sassi S, Triki H, Belghith M, Rezig D. Profile of Cytokines and T Cell Subsets Transcription Factors in Cerebrospinal Fluid of Patients with Viral Encephalitis. Viral Immunol 2024; 37:459-469. [PMID: 39527011 DOI: 10.1089/vim.2024.0058] [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] [Indexed: 11/16/2024] Open
Abstract
This study investigates the demographic, clinical characteristics, virological profiles, and immunological responses of patients with viral encephalitis (VE) compared with a control group. The VE group displayed a wide range of neurological symptoms. Virological analysis revealed the predominance of Herpesviridae family viruses. Immune responses in cerebrospinal fluid (CSF) from patients with VE were examined, highlighting an immunological shift toward T helper 1 (Th1) cells dominance, altered T helper 17 cells/regulatory T cells (Th17/Tregs) balance, and high interleukin-6 expression. These findings provide insights into the complex immunological landscape of VE, highlighting the role of specific cytokines and T cell subsets in its pathogenesis and potentially guiding targeted therapeutic strategies.
Collapse
Affiliation(s)
- Khadija Tebibi
- Research Laboratory "Virus, Vectors and Hosts: One Health Approach and Technological Innovation for a Better Health", Pasteur Institute of Tunis, Tunis, Tunisia
- Faculty of Sciences of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Rafika Ben Laamari
- Faculty of Sciences of Tunis, University of Tunis El Manar, Tunis, Tunisia
- Laboratory of Transmission, Control, and Immunobiology of Infections, Pasteur Institute of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Zakaria Saied
- Neurological Department of Mongi Ben Hmida Institute, Tunis, Tunisia
- Faculty of Medicine of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Olfa Maghrebi
- Laboratory of Transmission, Control, and Immunobiology of Infections, Pasteur Institute of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Henda Touzi
- Laboratory of Clinical Virology, WHO Reference Laboratory for Poliomyelitis and Measles in the Eastern Mediterranean Region, Pasteur Institute of Tunis, University of Tunis El Manar (UTM), Tunis, Tunisia
| | - Zina Meddeb
- Laboratory of Clinical Virology, WHO Reference Laboratory for Poliomyelitis and Measles in the Eastern Mediterranean Region, Pasteur Institute of Tunis, University of Tunis El Manar (UTM), Tunis, Tunisia
| | - Samia Ben Sassi
- Neurological Department of Mongi Ben Hmida Institute, Tunis, Tunisia
- Faculty of Medicine of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Henda Triki
- Research Laboratory "Virus, Vectors and Hosts: One Health Approach and Technological Innovation for a Better Health", Pasteur Institute of Tunis, Tunis, Tunisia
- Laboratory of Clinical Virology, WHO Reference Laboratory for Poliomyelitis and Measles in the Eastern Mediterranean Region, Pasteur Institute of Tunis, University of Tunis El Manar (UTM), Tunis, Tunisia
| | - Meriam Belghith
- Laboratory of Transmission, Control, and Immunobiology of Infections, Pasteur Institute of Tunis, University of Tunis El Manar, Tunis, Tunisia
| | - Dorra Rezig
- Research Laboratory "Virus, Vectors and Hosts: One Health Approach and Technological Innovation for a Better Health", Pasteur Institute of Tunis, Tunis, Tunisia
- Laboratory of Clinical Virology, WHO Reference Laboratory for Poliomyelitis and Measles in the Eastern Mediterranean Region, Pasteur Institute of Tunis, University of Tunis El Manar (UTM), Tunis, Tunisia
| |
Collapse
|
48
|
Prompsy P, Saichi M, Raimundo F, Vallot C. IDclust: Iterative clustering for unsupervised identification of cell types with single cell transcriptomics and epigenomics. NAR Genom Bioinform 2024; 6:lqae174. [PMID: 39703425 PMCID: PMC11655290 DOI: 10.1093/nargab/lqae174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/08/2024] [Accepted: 11/19/2024] [Indexed: 12/21/2024] Open
Abstract
The increasing diversity of single-cell datasets require systematic cell type characterization. Clustering is a critical step in single-cell analysis, heavily influencing downstream analyses. However, current unsupervised clustering algorithms rely on biologically irrelevant parameters that require manual optimization and fail to capture hierarchical relationships between clusters. We developed IDclust, a framework that identifies clusters with significant biological features at multiple resolutions using biologically meaningful thresholds like fold change, adjusted P-value and fraction of expressing cells. By iteratively processing and clustering subsets of the dataset, IDclust guarantees that all clusters found have significantly different features and stops only when no more interpretable cluster is found. It also creates a hierarchy of clusters, enabling visualization of the hierarchical relationships between different clusters. Analyzing multiple single-cell transcriptomic reference datasets, IDclust achieves superior clustering accuracy compared to state of the art algorithms. We showcase its utility by identifying previously unannotated clusters and identifying branching patterns in scATAC datasets. Using it's unsupervised nature and ability to analyze different -omics, we compare the resolution of different histone marks in multi-omic paired-tag dataset. Overall, IDclust automates single-cell exploration, facilitates cell type annotation and provides a biologically interpretable basis for clustering.
Collapse
Affiliation(s)
- Pacôme Prompsy
- CNRS UMR3244, Institut Curie, PSL Research University, 26 rue d’Ulm, 75005 Paris, France
- Department of Translational Research, Institut Curie, PSL Research University, 26 rue d’Ulm, 75005 Paris, France
- Department of Dermatology, Lausanne University Hospital (CHUV), Avenue de Beaumont 29, 1011Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Rue du Bugnon 46, 1005 Lausanne, Switzerland
| | - Mélissa Saichi
- CNRS UMR3244, Institut Curie, PSL Research University, 26 rue d’Ulm, 75005 Paris, France
- Department of Translational Research, Institut Curie, PSL Research University, 26 rue d’Ulm, 75005 Paris, France
| | - Félix Raimundo
- CNRS UMR3244, Institut Curie, PSL Research University, 26 rue d’Ulm, 75005 Paris, France
- Department of Translational Research, Institut Curie, PSL Research University, 26 rue d’Ulm, 75005 Paris, France
- Department of Genomics and Computational Biology, University of Massachusetts Chan Medical School, 55 N Lake Ave, Worcester, MA 01605, USA
| | - Céline Vallot
- CNRS UMR3244, Institut Curie, PSL Research University, 26 rue d’Ulm, 75005 Paris, France
- Department of Translational Research, Institut Curie, PSL Research University, 26 rue d’Ulm, 75005 Paris, France
| |
Collapse
|
49
|
Tao Y, Jiang Z, Wang H, Li J, Li X, Ni J, Liu J, Xiang H, Guan C, Cao W, Li D, He K, Wang L, Hu J, Jin Y, Liao B, Zhang T, Wu X. Pseudokinase STK40 promotes T H1 and T H17 cell differentiation by targeting FOXO transcription factors. SCIENCE ADVANCES 2024; 10:eadp2919. [PMID: 39565845 PMCID: PMC11578171 DOI: 10.1126/sciadv.adp2919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 10/18/2024] [Indexed: 11/22/2024]
Abstract
Inappropriate CD4+ T helper (TH) cell differentiation leads to progression of inflammatory and autoimmune diseases, yet the regulatory mechanisms governing stability and activity of transcription factors controlling TH cell differentiation remain elusive. Here, we describe how pseudokinase serine threonine kinase 40 (STK40) facilitates TH1/TH17 differentiation under pathological conditions. STK40 in T cells is dispensable for immune homeostasis in resting mice. However, mice with T cell-specific deletion of STK40 exhibit attenuated symptoms of experimental autoimmune encephalomyelitis and colitis, accompanied by diminished TH1 and TH17 cell differentiation. Mechanistically, STK40 facilitates K48-linked polyubiquitination and proteasomal degradation of FOXO1/4 through promoting their interaction with E3 ligase COP1. Inhibition of FOXO4 or FOXO1, respectively, restores differentiation potential of STK40-deficient TH1/TH17 cells. Together, our data suggest a crucial role of STK40 in TH1 and TH17 cell differentiation, thereby enabling better understanding of the molecular regulatory network of CD4+ T cell differentiation and providing effective targets for the treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Yuexiao Tao
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine; Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Shanghai Jiao Tong University School of Medicine; Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine; and Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenyan Jiang
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine; Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Shanghai Jiao Tong University School of Medicine; Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine; and Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huizi Wang
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine; Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Shanghai Jiao Tong University School of Medicine; Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine; and Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jia Li
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine; Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Shanghai Jiao Tong University School of Medicine; Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine; and Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xin Li
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine; Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Shanghai Jiao Tong University School of Medicine; Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine; and Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Ni
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine; Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Shanghai Jiao Tong University School of Medicine; Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine; and Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiamin Liu
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine; Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Shanghai Jiao Tong University School of Medicine; Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine; and Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hongrui Xiang
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine; Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Shanghai Jiao Tong University School of Medicine; Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine; and Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chenyang Guan
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine; Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Shanghai Jiao Tong University School of Medicine; Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine; and Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Cao
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine; Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Shanghai Jiao Tong University School of Medicine; Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine; and Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dongyang Li
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine; Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Shanghai Jiao Tong University School of Medicine; Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine; and Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ke He
- Department of Histoembryology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Lina Wang
- Department of Histoembryology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jing Hu
- Department of Histoembryology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ying Jin
- Department of Histoembryology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai 200031, China
| | - Bing Liao
- Department of Histoembryology, Genetics and Developmental Biology, Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ting Zhang
- Department of Gastroenterology, Hepatology, and Nutrition, Shanghai Children’s Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xuefeng Wu
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine; Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Shanghai Jiao Tong University School of Medicine; Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine; and Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
50
|
Leonard MR, Jones DM, Read KA, Pokhrel S, Tuazon JA, Warren RT, Yount JS, Oestreich KJ. Aiolos promotes CXCR3 expression on Th1 cells via positive regulation of IFN-γ/STAT1 signaling. JCI Insight 2024; 10:e180287. [PMID: 39560988 PMCID: PMC11721307 DOI: 10.1172/jci.insight.180287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 11/13/2024] [Indexed: 11/20/2024] Open
Abstract
CD4+ T helper 1 (Th1) cells coordinate adaptive immune responses to intracellular pathogens, including viruses. Key to this function is the ability of Th1 cells to migrate within secondary lymphoid tissues, as well as to sites of inflammation, which relies on signals received through the chemokine receptor CXCR3. CXCR3 expression is driven by the Th1 lineage-defining transcription factor T-bet and the cytokine-responsive STAT family members STAT1 and STAT4. Here, we identify the Ikaros zinc finger (IkZF) transcription factor Aiolos (Ikzf3) as an additional positive regulator of CXCR3 both in vitro and in vivo using a murine model of influenza virus infection. Mechanistically, we found that Aiolos-deficient CD4+ T cells exhibited decreased expression of key components of the IFN-γ/STAT1 signaling pathway, including JAK2 and STAT1. Consequently, Aiolos deficiency resulted in decreased levels of STAT1 tyrosine phosphorylation and reduced STAT1 enrichment at the Cxcr3 promoter. We further found that Aiolos and STAT1 formed a positive feedback loop via reciprocal regulation of each other downstream of IFN-γ signaling. Collectively, our study demonstrates that Aiolos promotes CXCR3 expression on Th1 cells by propagating the IFN-γ/STAT1 cytokine signaling pathway.
Collapse
Affiliation(s)
- Melissa R. Leonard
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, Ohio, USA
- Combined Anatomic Pathology Residency/PhD Program, The Ohio State University College of Veterinary Medicine, Columbus, Ohio, USA
| | - Devin M. Jones
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, Ohio, USA
- Biomedical Sciences Graduate Program and
| | - Kaitlin A. Read
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, Ohio, USA
- Biomedical Sciences Graduate Program and
| | - Srijana Pokhrel
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, Ohio, USA
| | - Jasmine A. Tuazon
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, Ohio, USA
- Biomedical Sciences Graduate Program and
- Medical Scientist Training Program, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Robert T. Warren
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, Ohio, USA
| | - Jacob S. Yount
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, Ohio, USA
- Infectious Diseases Institute, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, Ohio, USA
| | - Kenneth J. Oestreich
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, Ohio, USA
- Infectious Diseases Institute, The Ohio State University College of Medicine and Wexner Medical Center, Columbus, Ohio, USA
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA
| |
Collapse
|