1
|
Li G, Zhou Q, Xie M, Zhao B, Zhang K, Luo Y, Kong L, Gao D, Guo Y. Identification of ageing-associated gene signatures in heart failure with preserved ejection fraction by integrated bioinformatics analysis and machine learning. Genes Dis 2025; 12:101478. [PMID: 40330147 PMCID: PMC12053710 DOI: 10.1016/j.gendis.2024.101478] [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: 03/09/2024] [Revised: 10/10/2024] [Accepted: 11/21/2024] [Indexed: 05/08/2025] Open
Abstract
The incidence of heart failure with preserved ejection fraction (HFpEF) increases with the ageing of populations. This study aimed to explore ageing-associated gene signatures in HFpEF to develop new diagnostic biomarkers and provide new insights into the underlying mechanisms of HFpEF. Mice were subjected to a high-fat diet combined with L-NG-nitroarginine methyl ester (l-NAME) to induce HFpEF, and next-generation sequencing was performed with HFpEF hearts. Additionally, separate datasets were acquired from the Gene Expression Omnibus (GEO) database. The differentially expressed genes (DEGs) were used to identify ageing-related DEGs. Support vector machine, random forest, and least absolute shrinkage and selection operator algorithms were employed to identify potential diagnostic genes from ageing-related DEGs. The diagnostic value was assessed using a nomogram and receiver operating characteristic curve. The gene and related protein expression were verified by reverse transcription PCR and western blotting. The immune cell infiltration in hearts was analysed using the single-sample gene-set enrichment analysis algorithm. The results showed that the merged HFpEF datasets comprised 103 genes, of which 15 ageing-related DEGs were further screened in. The ageing-related DEGs were primarily associated with immune and metabolism regulation. AGTR1a, NR3C1, and PRKAB1 were selected for nomogram construction and machine learning-based diagnostic value, displaying strong diagnostic potential. Additionally, ageing scores were established based on nine key DEGs, revealing noteworthy differences in immune cell infiltration across HFpEF subtypes. In summary, those results highlight the significance of immune dysfunction in HFpEF. Furthermore, ageing-related DEGs might serve as promising prognostic and predictive biomarkers for HFpEF.
Collapse
Affiliation(s)
- Guoxing Li
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Cardiovascular Disease Laboratory of Chongqing Medical University, Chongqing 400016, China
| | - Qingju Zhou
- Department of Health Management Center, Chongqing General Hospital, Chongqing University, Chongqing 400010, China
| | - Ming Xie
- Department of Cardiothoracic Surgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University, Chongqing 400010, China
| | - Boying Zhao
- Department of Cardiothoracic Surgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University, Chongqing 400010, China
| | - Keyu Zhang
- Cardiovascular Disease Laboratory of Chongqing Medical University, Chongqing 400016, China
- Department of Vascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yuan Luo
- Department of Cardiothoracic Surgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University, Chongqing 400010, China
| | - Lingwen Kong
- Department of Cardiothoracic Surgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University, Chongqing 400010, China
| | - Diansa Gao
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Cardiovascular Disease Laboratory of Chongqing Medical University, Chongqing 400016, China
| | - Yongzheng Guo
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
- Cardiovascular Disease Laboratory of Chongqing Medical University, Chongqing 400016, China
- Department of Cardiothoracic Surgery, Chongqing Emergency Medical Center, Chongqing University Central Hospital, Chongqing University, Chongqing 400010, China
| |
Collapse
|
2
|
Li R, He T, Xing Z, Mi L, Su A, Wu W. The immune system in Hashimoto's thyroiditis: Updating the current state of knowledge on potential therapies and animal model construction. Autoimmun Rev 2025; 24:103783. [PMID: 40037463 DOI: 10.1016/j.autrev.2025.103783] [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: 10/24/2024] [Revised: 02/12/2025] [Accepted: 02/24/2025] [Indexed: 03/06/2025]
Abstract
Hashimoto's thyroiditis (HT) is one of the most prevalent endocrine disorders worldwide, and it can occur in people of all ages, including children. HT has a multifactorial pathogenesis and develops after a combination of gene regulation, environmental modifiers, and infection triggers. Various coamplifying feedback chronic inflammatory systems are involved in immune mechanisms, including oxidative stress, lymphocyte infiltration, and thyroid cell death. Furthermore, there is no effective treatment for HT at their roots. Thus, this review systematically summarizes and updates the existing etiology and pathogenesis, potential malignant transformation, emerging therapeutic drugs and animal model construction, making it more convenient for researchers to obtain effective information about HT and better explore potential strategies for short-term treatment of the disease.
Collapse
Affiliation(s)
- Ruixi Li
- Division of Thyroid Surgery, Department of General Surgery and Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ting He
- Division of Thyroid Surgery, Department of General Surgery and Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhichao Xing
- Division of Thyroid Surgery, Department of General Surgery and Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Li Mi
- Division of Thyroid Surgery, Department of General Surgery and Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Anping Su
- Division of Thyroid Surgery, Department of General Surgery and Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wenshuang Wu
- Division of Thyroid Surgery, Department of General Surgery and Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
3
|
Kakh M, Doroudchi M, Talepoor A. Induction of Regulatory T Cells After Virus Infection and Vaccination. Immunology 2025. [PMID: 40329764 DOI: 10.1111/imm.13927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 03/04/2025] [Accepted: 03/17/2025] [Indexed: 05/08/2025] Open
Abstract
Vaccines have been proven to be one of the safest and most effective ways to prevent and combat diseases. However, the main focus has been on the evaluation of the potency of effector mechanisms and the lack of adverse effects of vaccine candidates. Recently, the importance of induced regulatory mechanisms of the immune system after vaccination has come to light. With the increase in our knowledge about these regulatory mechanisms including the regulatory T cells (Tregs), we have come to understand the significance of this arm of the immune system in controlling immunopathology and/or diminishing the effectiveness of vaccines, especially viral vaccines. Tregs play a dual role during infectious diseases by limiting immune-mediated pathology and also contributing to chronic pathogen persistence by decreasing effector immunity and clearance of infection. Tregs may also affect immune responses after vaccination primarily by inhibiting antigen presenting cell function such as cytokine secretion and co-stimulatory molecule expression as well as effector T (Teff) and B cell function. In this article, we review the current knowledge on the induction of Tregs after several life-threatening virus infections and their available vaccines to bring them to the spotlight and emphasise that studying viral-induced antigen-specific Tregs will help us improve the effectiveness and decrease the immunopathology or side effects of viral vaccines. Trial Registration: ClinicalTrials.gov identifier: NCT04357444.
Collapse
Affiliation(s)
- MansourehKarimi Kakh
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehrnoosh Doroudchi
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - AtefeGhamar Talepoor
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Autoimmune Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
4
|
Tao K, Tao K, Wang J. The potential mechanisms of extracellular vesicles in transfusion-related adverse reactions: Recent advances. Transfus Clin Biol 2025; 32:205-227. [PMID: 40180029 DOI: 10.1016/j.tracli.2025.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/15/2025] [Accepted: 03/25/2025] [Indexed: 04/05/2025]
Abstract
Blood transfusion is an irreplaceable clinical treatment. Blood components are differentiated and stored according to specific guidelines. Storage temperatures and times vary depending on the blood component, but they all release extracellular vesicles (EVs) during storage. Although blood transfusions can be life-saving, they can also cause many adverse transfusion reactions, among which the effects of EVs are of increasing interest to researchers. EVs are submicron particles that vary in size, composition, and surface biomarkers, are encapsulated by a lipid bilayer, and are not capable of self-replication. EVs released by blood cells are important contributors to pathophysiologic states through proinflammatory, coagulant, and immunosuppressive effects, which in turn promote or inhibit the associated disease phenotype. Therefore, this review explores the potential mechanisms of hematopoietic-derived EVs in transfusion-associated adverse reactions and discusses the potential of the latest proteomics tools to be applied to the analysis of EVs in the field of transfusion medicine with a view to reducing the risk of blood transfusion.
Collapse
Affiliation(s)
- Keyi Tao
- Panzhihua University, Panzhihua 617000 Sichuan, China
| | - Keran Tao
- Institute of Medicine and Nursing, Hubei University of Medicine, Shiyan 442000 Hubei, China
| | - Jing Wang
- Southwest Medical University, Luzhou 646000 Sichuan, China; Department of Blood Transfusion, The Affiliated Hospital of Southwest Medical University, Luzhou Sichuan, 646000 China.
| |
Collapse
|
5
|
Xin M, Wang A, Ji M, Wu J, Jiang B, Shi M, Song L, Xin Z. Molecular Biology and Functions of T Follicular Helper Cells in Cancer and Immunotherapy. Immune Netw 2025; 25:e7. [PMID: 40342840 PMCID: PMC12056291 DOI: 10.4110/in.2025.25.e7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 12/22/2024] [Accepted: 01/08/2025] [Indexed: 05/11/2025] Open
Abstract
T follicular helper (Tfh) cells are integral to the germinal center (GC) response and the development of potent humoral immunity. By priming B cells, Tfh cells can initiate both extrafollicular and GC-dependent Ab responses. The dynamic physical interactions between Tfh and B cells constitute the primary platform for Tfh cells to provide essential "help" factors to B cells, as well as for reciprocal signaling from B cells to sustain the helper state of Tfh cells. In recent years, significant advancements have been made in understanding the diverse roles of Tfh cells across various diseases, particularly in cancer. Notably, beyond the classical GC-Tfh cells, it is increasingly recognized that the Tfh cell phenotype is highly heterogeneous and dynamic, which adds complexity to their roles in disease contexts. This review aims to encapsulate progress in Tfh cell biology, with a focus on their role in cancer and immunotherapy.
Collapse
Affiliation(s)
- Mengyuan Xin
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan 250117, China
| | - Antuo Wang
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan 250117, China
| | - Minghao Ji
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Jingru Wu
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Bin Jiang
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Mo Shi
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Liang Song
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Zhongwei Xin
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan 250117, China
| |
Collapse
|
6
|
Moura T, Laranjeira P, Caramelo O, Gil AM, Paiva A. Breast Cancer and Tumor Microenvironment: The Crucial Role of Immune Cells. Curr Oncol 2025; 32:143. [PMID: 40136347 PMCID: PMC11941043 DOI: 10.3390/curroncol32030143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/27/2025] Open
Abstract
Breast cancer is the most common type of cancer in women and the second leading cause of death by cancer. Despite recent advances, the mortality rate remains high, underlining the need to develop new therapeutic approaches. The complex interaction between cancer cells and the tumor microenvironment (TME) is crucial in determining tumor progression, therapy response, and patient prognosis. Understanding the role of immune cells in carcinogenesis and tumor progression can help improve targeted therapeutic options, increasing the likelihood of a favorable prognosis. Therefore, this review aims to critically analyze the complex interaction between tumor cells and immune cells, emphasizing the clinical and therapeutic implications. Additionally, we explore advances in immunotherapies, with a focus on immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Tânia Moura
- Flow Cytometry Unit, Department of Clinical Pathology, Hospitais da Universidade de Coimbra, Unidade Local de Saúde de Coimbra, 3000-076 Coimbra, Portugal; (T.M.); (P.L.)
- Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal;
| | - Paula Laranjeira
- Flow Cytometry Unit, Department of Clinical Pathology, Hospitais da Universidade de Coimbra, Unidade Local de Saúde de Coimbra, 3000-076 Coimbra, Portugal; (T.M.); (P.L.)
- Group of Environmental Genetics of Oncobiology (CIMAGO), Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3000-061 Coimbra, Portugal
- Center of Neurosciences and Cell (CNC), University of Coimbra, 3000-504 Coimbra, Portugal
| | - Olga Caramelo
- Gynecology Department, Hospitais da Universidade de Coimbra, Unidade Local de Saúde de Coimbra, 3000-075 Coimbra, Portugal;
| | - Ana M. Gil
- Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal;
- CICECO—Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Artur Paiva
- Flow Cytometry Unit, Department of Clinical Pathology, Hospitais da Universidade de Coimbra, Unidade Local de Saúde de Coimbra, 3000-076 Coimbra, Portugal; (T.M.); (P.L.)
- Group of Environmental Genetics of Oncobiology (CIMAGO), Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine (FMUC), University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3000-061 Coimbra, Portugal
- Ciências Biomédicas Laboratoriais, Instituto Politécnico de Coimbra, ESTESC—Coimbra Health School, 3046-854 Coimbra, Portugal
| |
Collapse
|
7
|
Sasaki T, Sowerby J, Xiao Y, Wang R, Marks KE, Horisberger A, Gao Y, Lee PY, Qu Y, Sze MA, Alves SE, Levesque MC, Fujio K, Costenbader KH, Rao DA. Clonal relationships between Tph and Tfh cells in patients with SLE and in murine lupus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.27.635189. [PMID: 39974998 PMCID: PMC11838332 DOI: 10.1101/2025.01.27.635189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Pathologic T cell-B cell interactions drive disease in systemic lupus erythematosus (SLE). The T cells that activate B cell responses include T peripheral helper (Tph) and T follicular helper (Tfh) cells, yet the developmental and clonal relationships between these B cell-helper T cell populations are unclear. Here we use T cell receptor (TCR) profiling to demonstrate clonal overlap between Tph and Tfh cells in the circulation of patients with SLE. Expanded Tph and Tfh cell clones persist over the course of 1 year in patients with a new diagnosis of SLE, and clones are observed to shift both from Tfh to Tph cells and from Tph to Tfh cells over time. High resolution analysis of cells sorted as Tph cells (CXCR5- PD-1hi) and Tfh cells (CXCR5+ PD-1hi) from SLE patients revealed considerable heterogeneity among cells sorted as Tph cells and highlighted a specific cluster of cells that expressed transcriptomic features of activated B cell-helper T cells. This cell population, marked by expression of TOX and CXCL13, was found in both sorted Tph and Tfh cells, and was clonally linked in these two populations. Analysis of B cell-helper T cells in murine pristane-induced lupus demonstrated similar populations of Tph and Tfh cells in both lung and spleen with strong clonal overlap. T cell-specific loss of Bcl6 prevented accumulation of Tfh cells and reduced accumulation of Tph cells in pristane-treated mice, indicating a role for Bcl6 in the survival and expansion of both populations. Together, these observations demonstrate a shared developmental path among pathologically expanded Tph and Tfh cells in lupus. The persistence of expanded Tph and Tfh cells clones over time may impose barriers to induction of stable tolerance by immunosuppressive medications or by B cell depletion.
Collapse
Affiliation(s)
- Takanori Sasaki
- Division of Rheumatology, Inflammation, Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - John Sowerby
- Division of Rheumatology, Inflammation, Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Yinan Xiao
- Division of Rheumatology, Inflammation, Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Runci Wang
- Division of Rheumatology, Inflammation, Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Kathryne E Marks
- Division of Rheumatology, Inflammation, Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Alice Horisberger
- Division of Rheumatology, Inflammation, Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Yidan Gao
- Division of Rheumatology, Inflammation, Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Pui Y Lee
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Yujie Qu
- Merck & Co., Inc., Boston, MA, USA
| | | | | | | | - Keishi Fujio
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Karen H Costenbader
- Division of Rheumatology, Inflammation, Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Deepak A Rao
- Division of Rheumatology, Inflammation, Immunity, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
8
|
Meléndez DC, Laniewski N, Jusko TA, Qiu X, Paige Lawrence B, Rivera-Núñez Z, Brunner J, Best M, Macomber A, Leger A, Kannan K, Miller RK, Barrett ES, O'Connor TG, Scheible K. In utero exposure to per - and polyfluoroalkyl substances (PFAS) associates with altered human infant T helper cell development. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.11.18.24317489. [PMID: 39606350 PMCID: PMC11601683 DOI: 10.1101/2024.11.18.24317489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Background Environmental exposures to chemical toxicants during gestation and infancy can dysregulate multiple developmental processes, causing lifelong effects. There is compelling evidence of PFAS-associated immunotoxicity in adults and children. However, the effect of developmental PFAS exposure on infant T-cell immunity is unreported, and, if present, could be implicated in immune-related health outcomes. Objectives We seek to model longitudinal changes in CD4+ T-cell subpopulations from birth through 12 months and their association with in-utero PFAS exposure and postnatal CD4+ T-cell frequencies and functions. Methods Maternal-infant dyads were recruited as part of the UPSIDE-ECHO cohort during the first trimester between 2015 and 2019 in Rochester, New York; dyads were followed through the infant's first birthday. Maternal PFAS concentrations (PFOS, PFOA, PFNA, and PFHXS) were quantified in serum during the second trimester using high-performance liquid chromatography and tandem mass spectrometry. Infant lymphocyte frequencies were assessed at birth, 6- and 12-months using mass cytometry and high-dimensional clustering methods. Linear mixed-effects models were employed to analyze the relationship between maternal PFAS concentrations and CD4+ T-cell subpopulations (n=200). All models included a PFAS and age interaction and were adjusted for parity, infant sex, and pre-pregnancy body mass index. Results In-utero PFAS exposure correlated with multiple CD4+ T-cell subpopulations in infants. The greatest effect sizes were seen in T-follicular helper (Tfh) and T-helper 2 (Th2) cells at 12 months. A log 2 -unit increase in PFOS was associated with lower Tfh [0.17% (95%CI: -0.30, -0.40)] and greater Th2 [0.27% (95%CI: 0.18, 0.35)] cell percentages at 12 months. Similar trends were observed for PFOA, PFNA, and PFHXS. Discussion Maternal PFAS exposures correlate with cell-specific changes in the infant T-cell compartment, including key CD4+ T-cell subpopulations that play central roles in coordinating well-regulated, protective immunity. Future studies into the role of PFAS-associated T-cell distribution and risk of adverse immune-related health outcomes in children are warranted.
Collapse
|
9
|
Liu J, Yue WL, Fan HZ, Luo YS, Feng GW, Li JF. Correlation of cTfh cells and memory B cells with AMR after renal transplantation. Transpl Immunol 2024; 86:102095. [PMID: 39038741 DOI: 10.1016/j.trim.2024.102095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 02/25/2024] [Accepted: 07/17/2024] [Indexed: 07/24/2024]
Abstract
Renal transplantation is the preferred treatment option for patients with end-stage renal disease (ESRD) in a clinical setting. Antibody mediated rejection (AMR) is one of the leading causes of graft dysfunction. To address the current shortcomings in the early diagnosis and treatment of AMR in clinical practice, this article analyzes the distribution of different circulating T follicular helper (cTfh) cell subtypes and B cell subpopulations in peripheral blood and detects the cytokine levels of chemokine ligand 13 (CXCL13), interleukin-21 (IL-21), and interleukin-4 (IL-4) related to cTfh cells in peripheral blood of kidney transplant recipients. Moreover, we also explore the correlation between cTfh cells, peripheral blood memory B cells, and AMR, their value as early predictive indicators of AMR, and explore potential therapeutic targets for AMR patients. Our results indicate that the proportion of cTfh cells increased at the onset of AMR, which plays an important role in antigen-specific B-cell immune regulation. Activation of cTfh cells in AMR patients correlates with phenotypes of memory B cells and plasma blasts. cTfh cells and memory B cells have promising diagnostic efficacies and predictive values for AMR. The proportion of cTfh cells to CD4+ T cells and the proportion of memory B cells to CD19+ B cells are correlated with serum creatinine levels, indicating that cTfh cells and memory B cells may be involved in the progression of AMR. In addition, the CXCL13, IL-21, and IL-4, which were associated with cTfh cells, may be involved in the onset of AMR.
Collapse
Affiliation(s)
- Jia Liu
- Henan Medical College, Dietetics Teaching and Research Section, Zhengzhou, China
| | - Wen-Long Yue
- The First Affiliated Hospital of Zhengzhou University, Department of Renal Transplantation, Zhengzhou, China
| | - Hong-Zhao Fan
- The First Affiliated Hospital of Zhengzhou University, Department of Renal Transplantation, Zhengzhou, China
| | - Yong-Sheng Luo
- The First Affiliated Hospital of Zhengzhou University, Department of Renal Transplantation, Zhengzhou, China
| | - Gui-Wen Feng
- The First Affiliated Hospital of Zhengzhou University, Department of Renal Transplantation, Zhengzhou, China.
| | - Jin-Feng Li
- The First Affiliated Hospital of Zhengzhou University, Department of Renal Transplantation, Zhengzhou, China.
| |
Collapse
|
10
|
Wang Y, Wang Q, He F, Qiao N, Li X, Wei L, Sun L, Dai W, Li Y, Pang X, Hu J, Huang C, Yang G, Pang C, Hu Z, Xing M, Wan C, Zhou D. Age-dependent decrease of circulating T follicular helper cells correlates with disease severity in elderly patients with COVID-19. Clin Immunol 2024; 266:110329. [PMID: 39067679 DOI: 10.1016/j.clim.2024.110329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 07/04/2024] [Accepted: 07/20/2024] [Indexed: 07/30/2024]
Abstract
Overwhelming evidence has shown that aging is a significant risk factor for COVID-19-related hospitalizations, death and other adverse health outcomes. Particular T cell subsets that susceptible to aging and associated with COVID-19 disease severity requires further elucidation. Our study recruited 57 elderly patients with acute COVID-19 and 27 convalescent donors. Adaptive immunity was assessed across the COVID-19 severity spectrum. Patients underwent age-dependent CD4+ T lymphopenia, preferential loss of circulating T follicular regulatory cells (cTfh) subsets including cTfh-em, cTfh-cm, cTfh1, cTfh2, cTfh17 and circulating T follicular regulatory cells (cTfr), which regulated antibody production through different pathways and correlated with COVID-19 severity, were observed. Moreover, vaccination improved cTfh-cm, cTfh2, cTfr proportion and promoted NAb production. In conclusion, the elderly had gone through age-dependent cTfh subsets deficiency, which impeded NAb production and enabled aggravation of COVID-19 to critical illness, whereas SARS-CoV-2 vaccine inoculation helped to rejuvenate cTfh, cTfr and intensify NAb responses.
Collapse
Affiliation(s)
- Yihan Wang
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, No. 22 Qixiangtai Road, Tianjin 300070, China
| | - Qiu Wang
- Department of Physical and Rehabilitation Medicine, Tianjin Medical University General Hospital, No. 154 Anshan Road, Tianjin 300052, China
| | - Furong He
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, No. 22 Qixiangtai Road, Tianjin 300070, China
| | - Nan Qiao
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, No. 22 Qixiangtai Road, Tianjin 300070, China
| | - Xuejun Li
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, No. 22 Qixiangtai Road, Tianjin 300070, China
| | - Liqun Wei
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, No. 22 Qixiangtai Road, Tianjin 300070, China
| | - Lingjin Sun
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, No. 22 Qixiangtai Road, Tianjin 300070, China
| | - Weiqian Dai
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, No. 22 Qixiangtai Road, Tianjin 300070, China
| | - Ying Li
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, No. 22 Qixiangtai Road, Tianjin 300070, China
| | - Xueyang Pang
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, No. 22 Qixiangtai Road, Tianjin 300070, China
| | - Jiayi Hu
- Department of Clinical Medicine, Tianjin Medical University, No. 22 Qixiangtai Road, Tianjin 300070, China
| | - Chuan Huang
- Department of Physical and Rehabilitation Medicine, Tianjin Medical University General Hospital, No. 154 Anshan Road, Tianjin 300052, China
| | - Guangchen Yang
- Department of Physical and Rehabilitation Medicine, Tianjin Medical University General Hospital, No. 154 Anshan Road, Tianjin 300052, China
| | - Chongjie Pang
- Department of Infectious Diseases, Tianjin Medical University General Hospital, No. 154 Anshan Road, Tianjin 300052, China
| | - Zhidong Hu
- Department of Clinical Laboratory, Tianjin Medical University General Hospital, No. 154 Anshan Road, Tianjin 300052, China
| | - Man Xing
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, No. 22 Qixiangtai Road, Tianjin 300070, China.
| | - Chunxiao Wan
- Department of Physical and Rehabilitation Medicine, Tianjin Medical University General Hospital, No. 154 Anshan Road, Tianjin 300052, China.
| | - Dongming Zhou
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, No. 22 Qixiangtai Road, Tianjin 300070, China; Shanghai Public Health Clinical Center, Fudan University, No. 2901 Caolang Road, Shanghai 201508, China.
| |
Collapse
|
11
|
Nelson CS, Sage PT. T FH cell responses endure in human lymph nodes after vaccination. Nat Immunol 2024; 25:1510-1512. [PMID: 39164478 DOI: 10.1038/s41590-024-01935-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2024]
Affiliation(s)
- Cody S Nelson
- Transplantation Research Center, Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Peter T Sage
- Transplantation Research Center, Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
12
|
Schattgen SA, Turner JS, Ghonim MA, Crawford JC, Schmitz AJ, Kim H, Zhou JQ, Awad W, Mettelman RC, Kim W, McIntire KM, Haile A, Klebert MK, Suessen T, Middleton WD, Teefey SA, Presti RM, Ellebedy AH, Thomas PG. Influenza vaccination stimulates maturation of the human T follicular helper cell response. Nat Immunol 2024; 25:1742-1753. [PMID: 39164477 PMCID: PMC11362011 DOI: 10.1038/s41590-024-01926-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 07/11/2024] [Indexed: 08/22/2024]
Abstract
The differentiation and specificity of human CD4+ T follicular helper cells (TFH cells) after influenza vaccination have been poorly defined. Here we profiled blood and draining lymph node (LN) samples from human volunteers for over 2 years after two influenza vaccines were administered 1 year apart to define the evolution of the CD4+ TFH cell response. The first vaccination induced an increase in the frequency of circulating TFH (cTFH) and LN TFH cells at week 1 postvaccination. This increase was transient for cTFH cells, whereas the LN TFH cells further expanded during week 2 and remained elevated in frequency for at least 3 months. We observed several distinct subsets of TFH cells in the LN, including pre-TFH cells, memory TFH cells, germinal center (GC) TFH cells and interleukin-10+ TFH cell subsets beginning at baseline and at all time points postvaccination. The shift toward a GC TFH cell phenotype occurred with faster kinetics after the second vaccine compared to the first vaccine. We identified several influenza-specific TFH cell clonal lineages, including multiple responses targeting internal influenza virus proteins, and found that each TFH cell state was attainable within a clonal lineage. Thus, human TFH cells form a durable and dynamic multitissue network.
Collapse
Affiliation(s)
- Stefan A Schattgen
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jackson S Turner
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Mohamed A Ghonim
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jeremy Chase Crawford
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Aaron J Schmitz
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Hyunjin Kim
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Julian Q Zhou
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Walid Awad
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Robert C Mettelman
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Wooseob Kim
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Microbiology, Korea University College of Medicine, Seoul, Korea
| | - Katherine M McIntire
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Alem Haile
- Clinical Trials Unit, Washington University School of Medicine, St. Louis, MO, USA
| | - Michael K Klebert
- Clinical Trials Unit, Washington University School of Medicine, St. Louis, MO, USA
| | - Teresa Suessen
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - William D Middleton
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Sharlene A Teefey
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Rachel M Presti
- Division of Infectious Diseases, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Ali H Ellebedy
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA.
- The Andrew M. and Jane M. Bursky Center for Human Immunology & Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA.
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St. Louis, MO, USA.
| | - Paul G Thomas
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
13
|
Russo C, Raiden S, Algieri S, Bruera MJ, De Carli N, Sarli M, Cairoli H, De Lillo L, Morales I, Seery V, Otero A, Sananez I, Simaz N, Alfiero G, Rubino G, Moya N, Aedo Portela L, Herrero M, Blanco M, Salcedo Pereira M, Ferrero F, Geffner J, Arruvito L. ATP-P2X7R pathway activation limits the Tfh cell compartment during pediatric RSV infection. Front Immunol 2024; 15:1397098. [PMID: 39044830 PMCID: PMC11263008 DOI: 10.3389/fimmu.2024.1397098] [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: 03/06/2024] [Accepted: 06/06/2024] [Indexed: 07/25/2024] Open
Abstract
Background Follicular helper T cells (Tfh) are pivotal in B cell responses. Activation of the purinergic receptor P2X7 on Tfh cells regulates their activity. We investigated the ATP-P2X7R axis in circulating Tfh (cTfh) cells during Respiratory Syncytial Virus (RSV) infection. Methods We analyzed two cohorts: children with RSV infection (moderate, n=30; severe, n=21) and healthy children (n=23). We utilized ELISA to quantify the levels of PreF RSV protein-specific IgG antibodies, IL-21 cytokine, and soluble P2X7R (sP2X7R) in both plasma and nasopharyngeal aspirates (NPA). Additionally, luminometry was employed to determine ATP levels in plasma, NPA and supernatant culture. The frequency of cTfh cells, P2X7R expression, and plasmablasts were assessed by flow cytometry. To evaluate apoptosis, proliferation, and IL-21 production by cTfh cells, we cultured PBMCs in the presence of Bz-ATP and/or P2X7R antagonist (KN-62) and a flow cytometry analysis was performed. Results In children with severe RSV disease, we observed diminished titers of neutralizing anti-PreF IgG antibodies. Additionally, severe infections, compared to moderate cases, were associated with fewer cTfh cells and reduced plasma levels of IL-21. Our investigation revealed dysregulation in the ATP-P2X7R pathway during RSV infection. This was characterized by elevated ATP levels in both plasma and NPA samples, increased expression of P2X7R on cTfh cells, lower levels of sP2X7R, and heightened ATP release from PBMCs upon stimulation, particularly evident in severe cases. Importantly, ATP exposure decreased cTfh proliferative response and IL-21 production, while promoting their apoptosis. The P2X7R antagonist KN-62 mitigated these effects. Furthermore, disease severity positively correlated with ATP levels in plasma and NPA samples and inversely correlated with cTfh frequency. Conclusion Our findings indicate that activation of the ATP-P2X7R pathway during RSV infection may contribute to limiting the cTfh cell compartment by promoting cell death and dysfunction, ultimately leading to increased disease severity.
Collapse
Affiliation(s)
- Constanza Russo
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA, Facultad de Medicina, Universidad de Buenos Aires- Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Silvina Raiden
- Departamento de Medicina, Hospital General de Niños Pedro de Elizalde, Buenos Aires, Argentina
| | - Silvia Algieri
- Servicio de Pediatría, Hospital Nacional Profesor Alejandro Posadas, Buenos Aires, Argentina
| | - María José Bruera
- Servicio de Pediatría, Hospital Nacional Profesor Alejandro Posadas, Buenos Aires, Argentina
| | - Norberto De Carli
- Servicio de Pediatría, Clínica del Niño de Quilmes, Buenos Aires, Argentina
| | - Mariam Sarli
- Servicio de Pediatría, Hospital Nacional Profesor Alejandro Posadas, Buenos Aires, Argentina
| | - Héctor Cairoli
- Departamento de Medicina, Hospital General de Niños Pedro de Elizalde, Buenos Aires, Argentina
| | - Leonardo De Lillo
- Departamento de Medicina, Hospital General de Niños Pedro de Elizalde, Buenos Aires, Argentina
| | - Ivanna Morales
- Departamento de Medicina, Hospital General de Niños Pedro de Elizalde, Buenos Aires, Argentina
| | - Vanesa Seery
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA, Facultad de Medicina, Universidad de Buenos Aires- Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Adrián Otero
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA, Facultad de Medicina, Universidad de Buenos Aires- Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Inés Sananez
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA, Facultad de Medicina, Universidad de Buenos Aires- Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Nancy Simaz
- Servicio de Pediatría, Hospital Nacional Profesor Alejandro Posadas, Buenos Aires, Argentina
| | - Gisela Alfiero
- Servicio de Pediatría, Hospital Nacional Profesor Alejandro Posadas, Buenos Aires, Argentina
| | - Gabriela Rubino
- Servicio de Pediatría, Hospital Nacional Profesor Alejandro Posadas, Buenos Aires, Argentina
| | - Néstor Moya
- Servicio de Pediatría, Hospital Nacional Profesor Alejandro Posadas, Buenos Aires, Argentina
| | - Luisa Aedo Portela
- Servicio de Pediatría, Clínica del Niño de Quilmes, Buenos Aires, Argentina
| | - Mauro Herrero
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA, Facultad de Medicina, Universidad de Buenos Aires- Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Marina Blanco
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA, Facultad de Medicina, Universidad de Buenos Aires- Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | | | - Fernando Ferrero
- Departamento de Medicina, Hospital General de Niños Pedro de Elizalde, Buenos Aires, Argentina
| | - Jorge Geffner
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA, Facultad de Medicina, Universidad de Buenos Aires- Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| | - Lourdes Arruvito
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA, Facultad de Medicina, Universidad de Buenos Aires- Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina
| |
Collapse
|
14
|
Hou Y, Cao Y, He Y, Dong L, Zhao L, Dong Y, Niu R, Bi Y, Liu G. SIRT3 Negatively Regulates TFH-Cell Differentiation in Cancer. Cancer Immunol Res 2024; 12:891-904. [PMID: 38630891 DOI: 10.1158/2326-6066.cir-23-0786] [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: 09/25/2023] [Revised: 01/22/2024] [Accepted: 04/09/2024] [Indexed: 04/19/2024]
Abstract
Follicular helper T (TFH) cells are essential for inducing germinal center (GC) reactions to mediate humoral adaptive immunity in tumors; however, the mechanisms underlying TFH-cell differentiation remain unclear. In this study, we found that the metabolism sensor sirtuin 3 (SIRT3) is critical for TFH-cell differentiation and GC formation during tumor development and viral infection. SIRT3 deficiency in CD4+ T cells intrinsically enhanced TFH-cell differentiation and GC reactions during tumor development and viral infection. Mechanistically, damaged oxidative phosphorylation (OXPHOS) compensatively triggered the NAD+-glycolysis pathway to provide a cellular energy supply, which was necessary for SIRT3 deficiency-induced TFH-cell differentiation. Blocking NAD+ synthesis-glycolysis signaling or recovering OXPHOS activities reversed the TFH-cell differentiation induced by SIRT3 deficiency. Moreover, the mTOR and hypoxia-inducible factor 1α (HIF1α) signaling axis was found to be responsible for TFH-cell differentiation induced by SIRT3 deficiency. HIF1α directly interacted with and regulated the activity of the transcription factor Bcl6. Thus, our findings identify a cellular energy compensatory mechanism, regulated by the mitochondrial sensor SIRT3, that triggers NAD+-dependent glycolysis during mitochondrial OXPHOS injuries and an mTOR-HIF1α-Bcl6 pathway to reprogram TFH-cell differentiation. These data have implications for future cancer immunotherapy research targeting SIRT3 in T cells.
Collapse
Affiliation(s)
- Yueru Hou
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Yejin Cao
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Ying He
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Lin Dong
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Longhao Zhao
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Yingjie Dong
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Ruiying Niu
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China
| | - Yujing Bi
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
| | - Guangwei Liu
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, China
| |
Collapse
|
15
|
Liu Y, Wang X, Xu J, Xu Q, Xing J, Zou J, Wang S, Peng H. Circular RNA CircZNF644 Facilitates Circulating Follicular Helper T Cells Response in Patients with Graves' Disease. J Immunol Res 2024; 2024:9527268. [PMID: 38966668 PMCID: PMC11223900 DOI: 10.1155/2024/9527268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 12/13/2023] [Accepted: 06/13/2024] [Indexed: 07/06/2024] Open
Abstract
Aberrant accumulation of circulating follicular helper T cells (cTfh) has been found in the peripheral blood mononuclear cells (PBMCs) of Graves' disease (GD) patients. However, the underlying mechanism that contributes to the imbalance of cTfh cells remains unknown. Previously, studies described a GD-related circular RNAs (circRNAs)-circZNF644 that might be associated with cTfh cells. This study aimed to investigate the role of circZNF644 on cTfh cells in GD patients. Here, we found that circZNF644 was highly stable expression in the PBMCs of GD patients, which was positively correlated with the serum levels of TSH receptor autoantibodies (TRAb). Knockdown of circZNF644 caused a reduction of the proportion of cTfh cells in vitro. Mechanistically, circZNF644 served as a ceRNA for miR-29a-3p to promote ICOS expression, resulting in increased cTfh cells. In the PBMCs of GD patients, circZNF644 expression was positively correlated with ICOS expression and the percentage of cTfh cells, but negatively related to miR-29a-3p expression. Additionally, a strong relationship between circZNF644 and IL-21 was revealed in GD patients, and silencing of circZNF644 inhibited IL-21 expression. Our study elucidated that elevated expression of circZNF644 is a key feature in the development of GD and may contribute to the pathogenic role of cTfh cells in GD.
Collapse
Affiliation(s)
- Yingzhao Liu
- Department of EndocrinologyThe Affiliated People's Hospital of Jiangsu UniversityZhenjiang Medical School of Nanjing Medical University, Zhenjiang 212002, China
| | - Xuehua Wang
- Department of EndocrinologyThe Fourth Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Juan Xu
- Department of Critical Care MedicineThe Affiliated People's Hospital of Jiangsu UniversityZhenjiang Medical School of Nanjing Medical University, Zhenjiang 212002, China
| | - Qian Xu
- Department of EndocrinologyThe Affiliated People's Hospital of Jiangsu UniversityZhenjiang Medical School of Nanjing Medical University, Zhenjiang 212002, China
| | - Jie Xing
- Department of Laboratory MedicineThe Affiliated People's Hospital of Jiangsu UniversityZhenjiang Medical School of Nanjing Medical University, Zhenjiang 212002, China
| | - Junli Zou
- Department of EndocrinologyThe Affiliated People's Hospital of Jiangsu UniversityZhenjiang Medical School of Nanjing Medical University, Zhenjiang 212002, China
| | - Shengjun Wang
- Department of Laboratory MedicineThe Affiliated People's Hospital of Jiangsu UniversityZhenjiang Medical School of Nanjing Medical University, Zhenjiang 212002, China
| | - Huiyong Peng
- Department of Laboratory MedicineThe Affiliated People's Hospital of Jiangsu UniversityZhenjiang Medical School of Nanjing Medical University, Zhenjiang 212002, China
- Department of Genetic ToxicologyThe Key Laboratory of Modern Toxicology of Ministry of EducationCenter for Global HealthSchool of Public HealthNanjing Medical University, Nanjing 211100, China
| |
Collapse
|
16
|
Kumar S, Basto AP, Ribeiro F, Almeida SCP, Campos P, Peres C, Pulvirenti N, Al-Khalidi S, Kilbey A, Tosello J, Piaggio E, Russo M, Gama-Carvalho M, Coffelt SB, Roberts EW, Geginat J, Florindo HF, Graca L. Specialized Tfh cell subsets driving type-1 and type-2 humoral responses in lymphoid tissue. Cell Discov 2024; 10:64. [PMID: 38834551 DOI: 10.1038/s41421-024-00681-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 04/16/2024] [Indexed: 06/06/2024] Open
Abstract
Effective antibody responses are essential to generate protective humoral immunity. Different inflammatory signals polarize T cells towards appropriate effector phenotypes during an infection or immunization. Th1 and Th2 cells have been associated with the polarization of humoral responses. However, T follicular helper cells (Tfh) have a unique ability to access the B cell follicle and support the germinal center (GC) responses by providing B cell help. We investigated the specialization of Tfh cells induced under type-1 and type-2 conditions. We first studied homogenous Tfh cell populations generated by adoptively transferred TCR-transgenic T cells in mice immunized with type-1 and type-2 adjuvants. Using a machine learning approach, we established a gene expression signature that discriminates Tfh cells polarized towards type-1 and type-2 response, defined as Tfh1 and Tfh2 cells. The distinct signatures of Tfh1 and Tfh2 cells were validated against datasets of Tfh cells induced following lymphocytic choriomeningitis virus (LCMV) or helminth infection. We generated single-cell and spatial transcriptomics datasets to dissect the heterogeneity of Tfh cells and their localization under the two immunizing conditions. Besides a distinct specialization of GC Tfh cells under the two immunizations and in different regions of the lymph nodes, we found a population of Gzmk+ Tfh cells specific for type-1 conditions. In human individuals, we could equally identify CMV-specific Tfh cells that expressed Gzmk. Our results show that Tfh cells acquire a specialized function under distinct types of immune responses and with particular properties within the B cell follicle and the GC.
Collapse
Affiliation(s)
- Saumya Kumar
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Afonso P Basto
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
- CIISA - Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Lisboa, Portugal
- Laboratório Associado para Ciência Animal e Veterinária (AL4AnimalS), Lisbon, Portugal
| | - Filipa Ribeiro
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Silvia C P Almeida
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Patricia Campos
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Carina Peres
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, Lisboa, Portugal
| | | | - Sarwah Al-Khalidi
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
- Cancer Research UK Scotland Institute, Glasgow, UK
| | - Anna Kilbey
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
- Cancer Research UK Scotland Institute, Glasgow, UK
| | - Jimena Tosello
- Institut Curie, PSL Research University, INSERM U932, Paris, France
| | - Eliane Piaggio
- Institut Curie, PSL Research University, INSERM U932, Paris, France
| | - Momtchilo Russo
- Institute of Biomedical Sciences, Department of Immunology, University of Sao Paulo, Sao Paulo, Brazil
| | - Margarida Gama-Carvalho
- BioISI - Biosystems & Integrative Sciences Institute, Faculty of Sciences, University of Lisboa, Lisboa, Portugal
| | - Seth B Coffelt
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
- Cancer Research UK Scotland Institute, Glasgow, UK
| | - Ed W Roberts
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
- Cancer Research UK Scotland Institute, Glasgow, UK
| | - Jens Geginat
- Istituto Nazionale di Genetica Molecolare, Milano, Italy
- Università degli studi di Milano, DISCCO, Milano, Italy
| | - Helena F Florindo
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, Lisboa, Portugal
| | - Luis Graca
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.
- Instituto Gulbenkian de Ciência, Oeiras, Portugal.
| |
Collapse
|
17
|
Wang J, Lou Y, Wang S, Zhang Z, You J, Zhu Y, Yao Y, Hao Y, Liu P, Xu LX. IFNγ at the early stage induced after cryo-thermal therapy maintains CD4 + Th1-prone differentiation, leading to long-term antitumor immunity. Front Immunol 2024; 15:1345046. [PMID: 38827732 PMCID: PMC11140566 DOI: 10.3389/fimmu.2024.1345046] [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: 11/27/2023] [Accepted: 04/29/2024] [Indexed: 06/04/2024] Open
Abstract
Introduction Recently, more and more research illustrated the importance of inducing CD4+ T helper type (Th)-1 dominant immunity for the success of tumor immunotherapy. Our prior studies revealed the crucial role of CD4+ Th1 cells in orchestrating systemic and durable antitumor immunity, which contributes to the satisfactory outcomes of the novel cryo-thermal therapy in the B16F10 tumor model. However, the mechanism for maintaining the cryo-thermal therapy-mediated durable CD4+ Th1-dominant response remains uncovered. Additionally, cryo-thermal-induced early-stage CD4+ Th1-dominant T cell response showed a correlation with the favorable prognosis in patients with colorectal cancer liver metastasis (CRCLM). We hypothesized that CD4+ Th1-dominant differentiation induced during the early stage post cryo-thermal therapy would affect the balance of CD4+ subsets at the late phase. Methods To understand the role of interferon (IFN)-γ, the major effector of Th1 subsets, in maintaining long-term CD4+ Th1-prone polarization, B16F10 melanoma model was established in this study and a monoclonal antibody was used at the early stage post cryo-thermal therapy for interferon (IFN)-γ signaling blockade, and the influence on the phenotypic and functional change of immune cells was evaluated. Results IFNγ at the early stage after cryo-thermal therapy maintained long-lasting CD4+ Th1-prone immunity by directly controlling Th17, Tfh, and Tregs polarization, leading to the hyperactivation of Myeloid-derived suppressor cells (MDSCs) represented by abundant interleukin (IL)-1β generation, and thereby further amplifying Th1 response. Discussion Our finding emphasized the key role of early-phase IFNγ abundance post cryo-thermal therapy, which could be a biomarker for better prognosis after cryo-thermal therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Ping Liu
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Lisa X. Xu
- School of Biomedical Engineering and Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
18
|
Yao Z, Yang L, Yang X, Liu F, Fu B, Xiong J. Stimulator of interferon genes mediated immune senescence reveals the immune microenvironment and prognostic characteristics of bladder cancer. Heliyon 2024; 10:e28803. [PMID: 38707337 PMCID: PMC11066586 DOI: 10.1016/j.heliyon.2024.e28803] [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: 10/30/2023] [Revised: 03/21/2024] [Accepted: 03/25/2024] [Indexed: 05/07/2024] Open
Abstract
Background Studies have shown that the stimulator of interferon genes (STING) is critical in tumorigenesis, and development. This study aimed to investigate the immune profile and prognostic significance of STING-mediated immune senescence in bladder cancer (BLCA). Methods We identified differential genes between tumor and normal tissue based on the Cancer Genome Atlas database, and used consensus clustering to identify BLCA subtypes. The genes most associated with overall survival were screened by further analysis and used to construct risk models. Then, comparing the immune microenvironment, tumor mutational load (TMB), and microsatellite instability (MSI) scores between different risk groups. Eventually, a nomogram was constructed based on clinical information and risk scores. The model was validated using receiver operating curves (ROC) and calibration plots. Results We identified 160 differential genes, including 13 genes most associated with prognosis. Three subtypes of bladder cancer with different clinical and immunological features were identified. Immunotherapy was more likely to benefit the low-risk group, which had higher TMB and MSI scores. The nomogram was found to be highly predictive based on ROC analysis and calibration plots. Conclusion The risk model and nomogram not only predict the prognosis of BLCA patients but also can guide the treatment.
Collapse
Affiliation(s)
- Zhijun Yao
- Department of Urology, Hengyang Central Hospital, Hengyang, 421001, China
| | - Lin Yang
- Department of Urology, First Affiliated Hospital of Nanchang University, Nanchang, 330000, China
| | - Xiaorong Yang
- Department of Urology, First Affiliated Hospital of Nanchang University, Nanchang, 330000, China
| | - Fang Liu
- Department of Urology, First Affiliated Hospital of Nanchang University, Nanchang, 330000, China
| | - Bin Fu
- Department of Urology, First Affiliated Hospital of Nanchang University, Nanchang, 330000, China
| | - Jing Xiong
- Department of Urology, First Affiliated Hospital of Nanchang University, Nanchang, 330000, China
| |
Collapse
|
19
|
Wang M, Zhou J, Niu Q, Wang H. Mechanism of tacrolimus in the treatment of lupus nephritis. Front Pharmacol 2024; 15:1331800. [PMID: 38774214 PMCID: PMC11106426 DOI: 10.3389/fphar.2024.1331800] [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: 11/01/2023] [Accepted: 04/19/2024] [Indexed: 05/24/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disorder, with more than half of the patients developing lupus nephritis (LN), which significantly contributes to chronic kidney disease (CKD) and end-stage renal disease (ESRD). The treatment of lupus nephritis has always been challenging. Tacrolimus (TAC), an effective immunosuppressant, has been increasingly used in the treatment of LN in recent years. This review aims to explore the mechanisms of action of tacrolimus in treating LN. Firstly, we briefly introduce the pharmacological properties of tacrolimus, including its role as a calcineurin (CaN) inhibitor, exerting immunosuppressive effects by inhibiting T cell activation and cytokine production. Subsequently, we focus on various other immunomodulatory mechanisms of tacrolimus in LN therapy, including its effects on T cells, B cells, and immune cells in kidney. Particularly, we emphasize tacrolimus' regulatory effect on inflammatory mediators and its importance in modulating the Th1/Th2 and Th17/Treg balance. Additionally, we review its effects on actin cytoskeleton, angiotensin II (Ang II)-specific vascular contraction, and P-glycoprotein activity, summarizing its impacts on non-immune mechanisms. Finally, we summarize the efficacy and safety of tacrolimus in clinical studies and trials. Although some studies have shown significant efficacy of tacrolimus in treating LN, its safety remains a challenge. We outline the potential adverse reactions of long-term tacrolimus use and provide suggestions on effectively monitoring and managing these adverse reactions in clinical practice. In general, tacrolimus, as a novel immunosuppressant, holds promising prospects for treating LN. Of course, further research is needed to better understand its therapeutic mechanisms and ensure its safety and efficacy in clinical practice.
Collapse
Affiliation(s)
| | | | | | - Hongyue Wang
- Department of Nephrology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
20
|
Luo RG, Wu YF, Lu HW, Weng D, Xu JY, Wang LL, Zhang LS, Zhao CQ, Li JX, Yu Y, Jia XM, Xu JF. Th2-skewed peripheral T-helper cells drive B-cells in allergic bronchopulmonary aspergillosis. Eur Respir J 2024; 63:2400386. [PMID: 38514095 PMCID: PMC11096668 DOI: 10.1183/13993003.00386-2024] [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: 09/26/2023] [Accepted: 03/07/2024] [Indexed: 03/23/2024]
Abstract
INTRODUCTION Patients with allergic bronchopulmonary aspergillosis (ABPA) suffer from repeated exacerbations. The involvement of T-cell subsets remains unclear. METHODS We enrolled ABPA patients, asthma patients and healthy controls. T-helper type 1 (Th1), 2 (Th2) and 17 (Th17) cells, regulatory T-cells (Treg) and interleukin (IL)-21+CD4+T-cells in total or sorted subsets of peripheral blood mononuclear cells and ABPA bronchoalveolar lavage fluid (BALF) were analysed using flow cytometry. RNA sequencing of subsets of CD4+T-cells was done in exacerbated ABPA patients and healthy controls. Antibodies of T-/B-cell co-cultures in vitro were measured. RESULTS ABPA patients had increased Th2 cells, similar numbers of Treg cells and decreased circulating Th1 and Th17 cells. IL-5+IL-13+IL-21+CD4+T-cells were rarely detected in healthy controls, but significantly elevated in the blood of ABPA patients, especially the exacerbated ones. We found that IL-5+IL-13+IL-21+CD4+T-cells were mainly peripheral T-helper (Tph) cells (PD-1+CXCR5-), which also presented in the BALF of ABPA patients. The proportions of circulating Tph cells were similar among ABPA patients, asthma patients and healthy controls, while IL-5+IL-13+IL-21+ Tph cells significantly increased in ABPA patients. Transcriptome data showed that Tph cells of ABPA patients were Th2-skewed and exhibited signatures of follicular T-helper cells. When co-cultured in vitro, Tph cells of ABPA patients induced the differentiation of autologous B-cells into plasmablasts and significantly enhanced the production of IgE. CONCLUSION We identified a distinctly elevated population of circulating Th2-skewed Tph cells that induced the production of IgE in ABPA patients. It may be a biomarker and therapeutic target for ABPA.
Collapse
Affiliation(s)
- Rong-Guang Luo
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- Institute of Respiratory Medicine, School of Medicine, Tongji University, Shanghai, China
- These authors contributed equally
| | - Yi-Fan Wu
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- Institute of Respiratory Medicine, School of Medicine, Tongji University, Shanghai, China
- These authors contributed equally
| | - Hai-Wen Lu
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- Institute of Respiratory Medicine, School of Medicine, Tongji University, Shanghai, China
- These authors contributed equally
| | - Dong Weng
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- Institute of Respiratory Medicine, School of Medicine, Tongji University, Shanghai, China
- These authors contributed equally
| | - Jia-Yan Xu
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- Institute of Respiratory Medicine, School of Medicine, Tongji University, Shanghai, China
| | - Le-Le Wang
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- Institute of Respiratory Medicine, School of Medicine, Tongji University, Shanghai, China
| | - Li-Sha Zhang
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- Institute of Respiratory Medicine, School of Medicine, Tongji University, Shanghai, China
| | - Cai-Qi Zhao
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- Institute of Respiratory Medicine, School of Medicine, Tongji University, Shanghai, China
| | - Jian-Xiong Li
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- Institute of Respiratory Medicine, School of Medicine, Tongji University, Shanghai, China
| | - Yong Yu
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Xin-Ming Jia
- Clinical Medicine Scientific and Technical Innovation Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jin-Fu Xu
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
- Institute of Respiratory Medicine, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
21
|
Kasten-Jolly J, Lawrence DA. Cellular and Molecular Immunity to Influenza Viruses and Vaccines. Vaccines (Basel) 2024; 12:389. [PMID: 38675771 PMCID: PMC11154265 DOI: 10.3390/vaccines12040389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 03/29/2024] [Accepted: 04/01/2024] [Indexed: 04/28/2024] Open
Abstract
Immune responses to influenza (flu) antigens reflect memory of prior infections or vaccinations, which might influence immunity to new flu antigens. Memory of past antigens has been termed "original antigenic sin" or, more recently, "immune imprinting" and "seniority". We have researched a comparison between the immune response to live flu infections and inactivated flu vaccinations. A brief history of antibody generation theories is presented, culminating in new findings about the immune-network theory and suggesting that a network of clones exists between anti-idiotypic antibodies and T cell receptors. Findings regarding the 2009 pandemic flu strain and immune responses to it are presented, including memory B cells and conserved regions within the hemagglutinin protein. The importance of CD4+ memory T cells and cytotoxic CD8+ T cells responding to both infections and vaccinations are discussed and compared. Innate immune cells, like natural killer (NK) cells and macrophages, are discussed regarding their roles in adaptive immune responses. Antigen presentation via macroautophagy processes is described. New vaccines in development are mentioned along with the results of some clinical trials. The manuscript concludes with how repeated vaccinations are impacting the immune system and a sketch of what might be behind the imprinting phenomenon, including future research directions.
Collapse
Affiliation(s)
- Jane Kasten-Jolly
- Wadsworth Center, New York State Department of Health, Albany, NY 12208, USA;
| | - David A. Lawrence
- Wadsworth Center, New York State Department of Health, Albany, NY 12208, USA;
- Departments of Biomedical Science and Environmental Health Science, University at Albany School of Public Health, Rensselaer, NY 12144, USA
| |
Collapse
|
22
|
Iborra-Pernichi M, Ruiz García J, Velasco de la Esperanza M, Estrada BS, Bovolenta ER, Cifuentes C, Prieto Carro C, González Martínez T, García-Consuegra J, Rey-Stolle MF, Rupérez FJ, Guerra Rodriguez M, Argüello RJ, Cogliati S, Martín-Belmonte F, Martínez-Martín N. Defective mitochondria remodelling in B cells leads to an aged immune response. Nat Commun 2024; 15:2569. [PMID: 38519473 PMCID: PMC10960012 DOI: 10.1038/s41467-024-46763-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 03/08/2024] [Indexed: 03/25/2024] Open
Abstract
The B cell response in the germinal centre (GC) reaction requires a unique bioenergetic supply. Although mitochondria are remodelled upon antigen-mediated B cell receptor stimulation, mitochondrial function in B cells is still poorly understood. To gain a better understanding of the role of mitochondria in B cell function, here we generate mice with B cell-specific deficiency in Tfam, a transcription factor necessary for mitochondrial biogenesis. Tfam conditional knock-out (KO) mice display a blockage of the GC reaction and a bias of B cell differentiation towards memory B cells and aged-related B cells, hallmarks of an aged immune response. Unexpectedly, blocked GC reaction in Tfam KO mice is not caused by defects in the bioenergetic supply but is associated with a defect in the remodelling of the lysosomal compartment in B cells. Our results may thus describe a mitochondrial function for lysosome regulation and the downstream antigen presentation in B cells during the GC reaction, the dysruption of which is manifested as an aged immune response.
Collapse
Affiliation(s)
- Marta Iborra-Pernichi
- Program of Tissue and Organ Homeostasis, Centro de Biología Molecular "Severo Ochoa", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
- Intestinal Morphogenesis and Homeostasis Group, Area 3-Cancer, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Jonathan Ruiz García
- Program of Tissue and Organ Homeostasis, Centro de Biología Molecular "Severo Ochoa", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
- Intestinal Morphogenesis and Homeostasis Group, Area 3-Cancer, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - María Velasco de la Esperanza
- Program of Tissue and Organ Homeostasis, Centro de Biología Molecular "Severo Ochoa", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
- Intestinal Morphogenesis and Homeostasis Group, Area 3-Cancer, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Belén S Estrada
- Program of Tissue and Organ Homeostasis, Centro de Biología Molecular "Severo Ochoa", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
- Intestinal Morphogenesis and Homeostasis Group, Area 3-Cancer, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Elena R Bovolenta
- Program of Tissue and Organ Homeostasis, Centro de Biología Molecular "Severo Ochoa", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
- Intestinal Morphogenesis and Homeostasis Group, Area 3-Cancer, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Claudia Cifuentes
- Program of Interactions with the Environment, Centro de Biología Molecular "Severo Ochoa", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
| | - Cristina Prieto Carro
- Program of Interactions with the Environment, Centro de Biología Molecular "Severo Ochoa", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
| | - Tamara González Martínez
- Program of Tissue and Organ Homeostasis, Centro de Biología Molecular "Severo Ochoa", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
- Intestinal Morphogenesis and Homeostasis Group, Area 3-Cancer, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - José García-Consuegra
- Program of Physiological and Pathological Processes, Centro de Biología Molecular "Severo Ochoa", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
| | - María Fernanda Rey-Stolle
- Centre for Metabolomics and Bioanalysis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Francisco Javier Rupérez
- Centre for Metabolomics and Bioanalysis (CEMBIO), Facultad de Farmacia, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Milagros Guerra Rodriguez
- Electron Microscopy Facility, Centro de Biología Molecular "Severo Ochoa, " Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
| | - Rafael J Argüello
- Aix Marseille Univ, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Sara Cogliati
- Program of Physiological and Pathological Processes, Centro de Biología Molecular "Severo Ochoa", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
| | - Fernando Martín-Belmonte
- Program of Tissue and Organ Homeostasis, Centro de Biología Molecular "Severo Ochoa", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain
- Intestinal Morphogenesis and Homeostasis Group, Area 3-Cancer, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Nuria Martínez-Martín
- Program of Tissue and Organ Homeostasis, Centro de Biología Molecular "Severo Ochoa", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain.
- Intestinal Morphogenesis and Homeostasis Group, Area 3-Cancer, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain.
| |
Collapse
|
23
|
Deng B, Huang H, Deng L, Zhao Z, Liu M, Lin H, Wang X, Tian R, Tu X, Peng A, Liang E, Bao K, Zhou Y, Xu P, He M. Imbalance of T follicular helper cell subsets trigger the differentiation of pathogenic B cells in idiopathic membranous nephropathy. Inflamm Res 2024:10.1007/s00011-023-01838-5. [PMID: 38467875 DOI: 10.1007/s00011-023-01838-5] [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: 08/02/2023] [Revised: 10/19/2023] [Accepted: 12/12/2023] [Indexed: 03/13/2024] Open
Abstract
OBJECTIVE This study aims to elucidate the role of T follicular helper (Tfh) cells and their subsets in idiopathic membranous nephropathy (IMN). METHODS The frequencies of Tfh cell subsets and B cell subsets in peripheral blood (PB) were detected in both IMN patients and healthy controls (HCs). The involvement of Tfh cells in the disease pathogenesis was examined by coculturing human Tfh cells with B cells. The dynamic changes of Tfh cells in PB or spleen were monitored in passive Heymann nephritis (PHN) rats. RESULTS The frequencies of circulating Tfh (cTfh) cells, cTfh2 cells, and plasmablasts were enriched in the PB of patients with IMN. cTfh cells expressed higher ICOS, and lower BTLA than healthy counterparts. The frequency of ICOS + cTfh2 was associated with the severity of IMN, including 24h urine protein, IgG4 concentration and the IgG4: IgG ratio. Positive correlations were also observed between the frequency of cTfh2 cells with plasmablasts, serum IL-21 and IL-4 levels. Importantly, cTfh cells isolated from IMN patients were able to induce the differentiation of B cells to memory B cells (MBC) and plasmablasts, this process could be substantially attenuated by blocking the IL-21. Similar increases of ICOS + cTfh cells were also detected in spleen of PHN rats, concomitant with elevated urine protein levels. CONCLUSIONS Collectively, our results demonstrate that the imbalance of cTfh cell subsets play a crucial pathogenic role in IMN by inducing the differentiation of B cells through IL-21, and cTfh2 cells might serve as useful markers to evaluate the progression of IMN.
Collapse
Affiliation(s)
- Bishun Deng
- Department of Laboratory Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Huijie Huang
- Department of Laboratory Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Li Deng
- Department of Laboratory Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ziling Zhao
- Department of Laboratory Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Miao Liu
- Department of Laboratory Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Haibiao Lin
- Department of Laboratory Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaowan Wang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Nephrology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ruimin Tian
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Nephrology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaoxin Tu
- Department of Laboratory Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Anping Peng
- Department of Laboratory Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Enyu Liang
- Department of Laboratory Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Kun Bao
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Nephrology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuting Zhou
- Clinical Laboratory of Urumqi Blood Center, Urumqi, China
| | - Peng Xu
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.
- Department of Nephrology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.
| | - Min He
- Department of Laboratory Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China.
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
24
|
White CL, Glover MA, Gandhapudi SK, Richards KA, Sant AJ. Flublok Quadrivalent Vaccine Adjuvanted with R-DOTAP Elicits a Robust and Multifunctional CD4 T Cell Response That Is of Greater Magnitude and Functional Diversity Than Conventional Adjuvant Systems. Vaccines (Basel) 2024; 12:281. [PMID: 38543915 PMCID: PMC10975948 DOI: 10.3390/vaccines12030281] [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: 01/31/2024] [Revised: 02/20/2024] [Accepted: 02/29/2024] [Indexed: 04/01/2024] Open
Abstract
It is clear that new approaches are needed to promote broadly protective immunity to viral pathogens, particularly those that are prone to mutation and escape from antibody-mediated immunity. CD4+ T cells, known to target many viral proteins and highly conserved peptide epitopes, can contribute greatly to protective immunity through multiple mechanisms. Despite this potential, CD4+ T cells are often poorly recruited by current vaccine strategies. Here, we have analyzed a promising new adjuvant (R-DOTAP), as well as conventional adjuvant systems AddaVax with or without an added TLR9 agonist CpG, to promote CD4+ T cell responses to the licensed vaccine Flublok containing H1, H3, and HA-B proteins. Our studies, using a preclinical mouse model of vaccination, revealed that the addition of R-DOTAP to Flublok dramatically enhances the magnitude and functionality of CD4+ T cells specific for HA-derived CD4+ T cell epitopes, far outperforming conventional adjuvant systems based on cytokine EliSpot assays and multiparameter flow cytometry. The elicited CD4+ T cells specific for HA-derived epitopes produce IL-2, IFN-γ, IL-4/5, and granzyme B and have multifunctional potential. Hence, R-DOTAP, which has been verified safe by human studies, can offer exciting opportunities as an immune stimulant for next-generation prophylactic recombinant protein-based vaccines.
Collapse
Affiliation(s)
- Chantelle L. White
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester, Rochester, NY 14642, USA; (C.L.W.); (M.A.G.); (K.A.R.)
| | - Maryah A. Glover
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester, Rochester, NY 14642, USA; (C.L.W.); (M.A.G.); (K.A.R.)
| | - Siva K. Gandhapudi
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky School of Medicine, Lexington, KY 40508, USA;
| | - Katherine A. Richards
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester, Rochester, NY 14642, USA; (C.L.W.); (M.A.G.); (K.A.R.)
| | - Andrea J. Sant
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester, Rochester, NY 14642, USA; (C.L.W.); (M.A.G.); (K.A.R.)
| |
Collapse
|
25
|
Chang Y, Bach L, Hasiuk M, Wen L, Elmzzahi T, Tsui C, Gutiérrez-Melo N, Steffen T, Utzschneider DT, Raj T, Jost PJ, Heink S, Cheng J, Burton OT, Zeiträg J, Alterauge D, Dahlström F, Becker JC, Kastl M, Symeonidis K, van Uelft M, Becker M, Reschke S, Krebs S, Blum H, Abdullah Z, Paeschke K, Ohnmacht C, Neumann C, Liston A, Meissner F, Korn T, Hasenauer J, Heissmeyer V, Beyer M, Kallies A, Jeker LT, Baumjohann D. TGF-β specifies T FH versus T H17 cell fates in murine CD4 + T cells through c-Maf. Sci Immunol 2024; 9:eadd4818. [PMID: 38427718 DOI: 10.1126/sciimmunol.add4818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 09/20/2023] [Accepted: 01/05/2024] [Indexed: 03/03/2024]
Abstract
T follicular helper (TFH) cells are essential for effective antibody responses, but deciphering the intrinsic wiring of mouse TFH cells has long been hampered by the lack of a reliable protocol for their generation in vitro. We report that transforming growth factor-β (TGF-β) induces robust expression of TFH hallmark molecules CXCR5 and Bcl6 in activated mouse CD4+ T cells in vitro. TGF-β-induced mouse CXCR5+ TFH cells are phenotypically, transcriptionally, and functionally similar to in vivo-generated TFH cells and provide critical help to B cells. The study further reveals that TGF-β-induced CXCR5 expression is independent of Bcl6 but requires the transcription factor c-Maf. Classical TGF-β-containing T helper 17 (TH17)-inducing conditions also yield separate CXCR5+ and IL-17A-producing cells, highlighting shared and distinct cell fate trajectories of TFH and TH17 cells. We demonstrate that excess IL-2 in high-density T cell cultures interferes with the TGF-β-induced TFH cell program, that TFH and TH17 cells share a common developmental stage, and that c-Maf acts as a switch factor for TFH versus TH17 cell fates in TGF-β-rich environments in vitro and in vivo.
Collapse
Affiliation(s)
- Yinshui Chang
- Medical Clinic III for Oncology, Hematology, Immuno-Oncology and Rheumatology, University Hospital Bonn, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
- Institute for Immunology, Faculty of Medicine, Biomedical Center, LMU Munich, Grosshaderner Str. 9, 82152 Planegg-Martinsried, Germany
| | - Luisa Bach
- Medical Clinic III for Oncology, Hematology, Immuno-Oncology and Rheumatology, University Hospital Bonn, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Marko Hasiuk
- Department of Biomedicine, Basel University Hospital and University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland
- Transplantation Immunology and Nephrology, Basel University Hospital, Petersgraben 4, CH-4031 Basel, Switzerland
| | - Lifen Wen
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria 3000, Australia
| | - Tarek Elmzzahi
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria 3000, Australia
- Immunogenomics and Neurodegeneration, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
| | - Carlson Tsui
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria 3000, Australia
| | - Nicolás Gutiérrez-Melo
- Medical Clinic III for Oncology, Hematology, Immuno-Oncology and Rheumatology, University Hospital Bonn, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Teresa Steffen
- Medical Clinic III for Oncology, Hematology, Immuno-Oncology and Rheumatology, University Hospital Bonn, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Daniel T Utzschneider
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria 3000, Australia
| | - Timsse Raj
- Institute for Immunology, Faculty of Medicine, Biomedical Center, LMU Munich, Grosshaderner Str. 9, 82152 Planegg-Martinsried, Germany
| | - Paul Jonas Jost
- Faculty of Mathematics and Natural Sciences, University of Bonn, Bonn, Germany
| | - Sylvia Heink
- Institute for Experimental Neuroimmunology, Technical University of Munich School of Medicine, 81675 Munich, Germany
| | - Jingyuan Cheng
- Experimental Systems Immunology, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Oliver T Burton
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Julia Zeiträg
- Institute for Immunology, Faculty of Medicine, Biomedical Center, LMU Munich, Grosshaderner Str. 9, 82152 Planegg-Martinsried, Germany
| | - Dominik Alterauge
- Institute for Immunology, Faculty of Medicine, Biomedical Center, LMU Munich, Grosshaderner Str. 9, 82152 Planegg-Martinsried, Germany
| | - Frank Dahlström
- Institute for Immunology, Faculty of Medicine, Biomedical Center, LMU Munich, Grosshaderner Str. 9, 82152 Planegg-Martinsried, Germany
| | - Jennifer-Christin Becker
- Medical Clinic III for Oncology, Hematology, Immuno-Oncology and Rheumatology, University Hospital Bonn, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Melanie Kastl
- Medical Clinic III for Oncology, Hematology, Immuno-Oncology and Rheumatology, University Hospital Bonn, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
- Department of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Konstantinos Symeonidis
- Institute of Molecular Medicine and Experimental Immunology, University Hospital Bonn, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Martina van Uelft
- Genomics and Immunoregulation, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Matthias Becker
- Systems Medicine, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
- PRECISE Platform for Single Cell Genomics and Epigenomics, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) and the University of Bonn, Bonn, Germany
| | - Sarah Reschke
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, Feodor-Lynen-Str. 25, 81377 Munich, Germany
| | - Stefan Krebs
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, Feodor-Lynen-Str. 25, 81377 Munich, Germany
| | - Helmut Blum
- Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, Feodor-Lynen-Str. 25, 81377 Munich, Germany
| | - Zeinab Abdullah
- Institute of Molecular Medicine and Experimental Immunology, University Hospital Bonn, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Katrin Paeschke
- Medical Clinic III for Oncology, Hematology, Immuno-Oncology and Rheumatology, University Hospital Bonn, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
- Department of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
| | - Caspar Ohnmacht
- Center of Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, Munich, Germany
| | - Christian Neumann
- Department of Microbiology, Infectious Diseases and Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Adrian Liston
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Felix Meissner
- Experimental Systems Immunology, Max Planck Institute of Biochemistry, Martinsried, Germany
- Department of Systems Immunology and Proteomics, Institute of Innate Immunity, Medical Faculty, University of Bonn, Germany
| | - Thomas Korn
- Institute for Experimental Neuroimmunology, Technical University of Munich School of Medicine, 81675 Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Jan Hasenauer
- Faculty of Mathematics and Natural Sciences, University of Bonn, Bonn, Germany
- Center for Mathematics, Technical University of Munich, Garching, Germany
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Vigo Heissmeyer
- Institute for Immunology, Faculty of Medicine, Biomedical Center, LMU Munich, Grosshaderner Str. 9, 82152 Planegg-Martinsried, Germany
- Research Unit Molecular Immune Regulation, Helmholtz Zentrum München, Feodor-Lynen-Str. 21, 81377 Munich, Germany
| | - Marc Beyer
- Immunogenomics and Neurodegeneration, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
- PRECISE Platform for Single Cell Genomics and Epigenomics, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) and the University of Bonn, Bonn, Germany
| | - Axel Kallies
- The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria 3000, Australia
| | - Lukas T Jeker
- Department of Biomedicine, Basel University Hospital and University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland
- Transplantation Immunology and Nephrology, Basel University Hospital, Petersgraben 4, CH-4031 Basel, Switzerland
| | - Dirk Baumjohann
- Medical Clinic III for Oncology, Hematology, Immuno-Oncology and Rheumatology, University Hospital Bonn, University of Bonn, Venusberg-Campus 1, 53127 Bonn, Germany
- Institute for Immunology, Faculty of Medicine, Biomedical Center, LMU Munich, Grosshaderner Str. 9, 82152 Planegg-Martinsried, Germany
| |
Collapse
|
26
|
Zhang Z, Jiang Q, Huang L, Huang J, Wan Q, Zhong Y, Liu D, Zhou W, Zhao H. Ginsenoside Rg1 regulated subpopulation homeostasis of Tfh cells ameliorate experimental colitis by inhibiting TLR/MyD88 pathway. J Funct Foods 2024; 113:106011. [DOI: 10.1016/j.jff.2024.106011] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/20/2024] Open
|
27
|
DiToro D, Murakami N, Pillai S. T-B Collaboration in Autoimmunity, Infection, and Transplantation. Transplantation 2024; 108:386-398. [PMID: 37314442 PMCID: PMC11345790 DOI: 10.1097/tp.0000000000004671] [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] [Indexed: 06/15/2023]
Abstract
We have attempted here to provide an up-to-date review of the collaboration between helper T cells and B cells in response to protein and glycoprotein antigens. This collaboration is essential as it not only protects from many pathogens but also contributes to a litany of autoimmune and immune-mediated diseases.
Collapse
Affiliation(s)
- Daniel DiToro
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard, Cambridge, MA
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA
| | - Naoka Murakami
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Shiv Pillai
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard, Cambridge, MA
| |
Collapse
|
28
|
Ji S, Shi Y, Yin B. Macrophage barrier in the tumor microenvironment and potential clinical applications. Cell Commun Signal 2024; 22:74. [PMID: 38279145 PMCID: PMC10811890 DOI: 10.1186/s12964-023-01424-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 12/05/2023] [Indexed: 01/28/2024] Open
Abstract
The tumor microenvironment (TME) constitutes a complex microenvironment comprising a diverse array of immune cells and stromal components. Within this intricate context, tumor-associated macrophages (TAMs) exhibit notable spatial heterogeneity. This heterogeneity contributes to various facets of tumor behavior, including immune response modulation, angiogenesis, tissue remodeling, and metastatic potential. This review summarizes the spatial distribution of macrophages in both the physiological environment and the TME. Moreover, this paper explores the intricate interactions between TAMs and diverse immune cell populations (T cells, dendritic cells, neutrophils, natural killer cells, and other immune cells) within the TME. These bidirectional exchanges form a complex network of immune interactions that influence tumor immune surveillance and evasion strategies. Investigating TAM heterogeneity and its intricate interactions with different immune cell populations offers potential avenues for therapeutic interventions. Additionally, this paper discusses therapeutic strategies targeting macrophages, aiming to uncover novel approaches for immunotherapy. Video Abstract.
Collapse
Affiliation(s)
- Shuai Ji
- Department of Urinary Surgery, The Shengjing Hospital of China Medical University, Shenyang, 110022, China
| | - Yuqing Shi
- Department of Respiratory Medicine, Shenyang 10th People's Hospital, Shenyang, 110096, China
| | - Bo Yin
- Department of Urinary Surgery, The Shengjing Hospital of China Medical University, Shenyang, 110022, China.
| |
Collapse
|
29
|
Wu R, Wang D, Cheng L, Su R, Li B, Fan C, Gao C, Wang C. Impaired immune tolerance mediated by reduced Tfr cells in rheumatoid arthritis linked to gut microbiota dysbiosis and altered metabolites. Arthritis Res Ther 2024; 26:21. [PMID: 38218985 PMCID: PMC10787489 DOI: 10.1186/s13075-023-03260-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 12/28/2023] [Indexed: 01/15/2024] Open
Abstract
BACKGROUND Patients with rheumatoid arthritis (RA) showed impaired immune tolerance characterized by reduced follicular regulatory T (Tfr) cells, and they also exhibited altered gut microbiotas and their metabolites in RA. However, the association of gut microbiotas and their metabolites with the immune tolerance mediated by Tfr cells in RA remains unclear. METHODS Peripheral blood and stool samples were collected from 32 new-onset RA patients and 17 healthy controls (HCs) in the Second Hospital of Shanxi Medical University between January 2022 and June 2022. The peripheral blood was used to detect the circulating regulatory T (Treg), helper T(Th)17, Tfr, and follicular helper T (Tfh) cells by modified flow cytometry. The stool samples were used to analyze the gut microbiotas and their metabolites via 16S rDNA sequencing and metabolomic profiling. We aimed to characterize the gut microbiotas and their metabolites in RA and identified their association with Tfr cell-mediated immune tolerance. RESULTS The new-onset RA demonstrated reduced Treg and Tfr cells, associated with the disease activity and autoantibodies. There were significant differences in gut microbiotas between the two groups as the results of β diversity analysis (P = 0.039) including 21 differential gut microbiotas from the phylum to genus levels. In which, Ruminococcus 2 was associated with the disease activity and autoantibodies of RA, and it was identified as the potential biomarker of RA [area under curve (AUC) = 0.782, 95% confidence interval (CI) = 0.636-0.929, P = 0.001]. Eleven differential metabolites were identified and participated in four main pathways related to RA. Arachidonic acid might be the potential biomarker of RA (AUC = 0.724, 95% CI = 0.595-0.909, P = 0.038), and it was the core metabolite as the positive association with six gut microbiotas enriched in RA. The reduced Tfr cells were associated with the altered gut microbiotas and their metabolites including the Ruminococcus 2, the arachidonic acid involved in the biosynthesis of unsaturated fatty acid pathway and the 3-methyldioxyindole involved in the tryptophan metabolism pathway. CONCLUSION The breakdown of immune tolerance mediated by reduced Tfr cells was associated with the altered gut microbiotas and their metabolites implying the possible mechanism of RA pathogenesis from the perspective of microecology-metabolism-immune.
Collapse
Affiliation(s)
- Ruihe Wu
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Immunomicroecology, Taiyuan, Shanxi, China
| | - Dongming Wang
- Department of Orthopedics, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Liyun Cheng
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Immunomicroecology, Taiyuan, Shanxi, China
| | - Rui Su
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Immunomicroecology, Taiyuan, Shanxi, China
| | - Baochen Li
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Immunomicroecology, Taiyuan, Shanxi, China
| | - Chunxue Fan
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Key Laboratory of Immunomicroecology, Taiyuan, Shanxi, China
| | - Chong Gao
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Caihong Wang
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China.
- Shanxi Key Laboratory of Immunomicroecology, Taiyuan, Shanxi, China.
| |
Collapse
|
30
|
Orfanakis M, Molyvdas A, Petrovas C. In Situ Characterization of Human Follicular Helper CD4 T Cells. Methods Mol Biol 2024; 2813:281-293. [PMID: 38888784 DOI: 10.1007/978-1-0716-3890-3_18] [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: 06/20/2024]
Abstract
The development of an effective humoral response to pathogens and immunogens is a multiphase biological process, which is mediated by the coordinated function of specialized immune cell types in secondary lymphoid organs and particularly in T cell and follicular areas. More specifically, within the follicular/germinal center area, the orchestrated interplay between B cells, follicular helper CD4 T cells (Tfh), and stromal cells triggers a cascade of immune reactions leading to the development of memory B cells and plasma cells able to generate effective, antigen-specific antibodies. The role of Tfh cells in this process is critical. Given the need for vaccines capable to induce antibodies of high affinity, neutralizing activity, and durability, understanding the cellular and molecular mechanisms regulating Tfh cell development is of great importance. Here, we describe novel approaches for the comprehensive understanding of these cells and possible implications for future studies in vaccine development and the understanding of the pathogenesis of relevant diseases.
Collapse
Affiliation(s)
- Michail Orfanakis
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland
| | - Adam Molyvdas
- Tissue Analysis Core, Immunology Laboratory, Vaccine Research Center, NIAID, NIH, Bethesda, MD, USA
| | - Constantinos Petrovas
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital and Lausanne University, Lausanne, Switzerland.
| |
Collapse
|
31
|
Huang Y, Sun X, Nie G, Xu H, Zou M. Follicular Helper T Cells in Peyer's Patches and Galactose- Deficient Iga1 Contribute to Iga Nephropathy. Curr Mol Med 2024; 24:1033-1044. [PMID: 37475555 DOI: 10.2174/1566524023666230720112215] [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: 12/14/2022] [Revised: 06/05/2023] [Accepted: 06/05/2023] [Indexed: 07/22/2023]
Abstract
BACKGROUND Common primary glomerulonephritis with aberrant mucosal immunity is IgA nephropathy (IgAN). T follicular helper (TFH) cells are essential in regulating B cell differentiation. Peyer's patches (PPs) are the main site where IgA+ plasmablasts differentiate. OBJECTIVE Our study aimed to investigate the TFH cell's potential contribution to the etiology of IgA nephropathy. METHODS In PPs from IgAN mouse models, the ratio of the TFH cell, B220+IgA+, B220+IgM+, and B220-IgA+ lymphocytes were assessed. Then, we used Western blot to assess the expression of Bcl-6, Blimp- 1, and IL-21 proteins in PPs and used RTPCR to assess the expression of IL-21 and TGF-β1 mRNA. TFH cells coculture with spleen cells to measure the degree of IL-21 and the ratio of activation marker CD69 on the TFH cells. Naive B cells (CD27-IgD+) from children suffering from IgAN were cultured with TFH cell-related cytokines. The supernatant was detected to assess the excretion of galactose-deficient IgA1 (Gd-IgA1). RESULTS IgAN mice developed noticeably increased degrees of IL-21 and CD69 on TFH cells than controls did, as well as higher percentages of B220+IgA+, B220+IgM+, B220+IgA+, TGF- β1, and IL-21 mRNA and Bcl-6, IL-21 proteins in PPs. The Gd-IgA1 level in the supernatant and IgAN- positive children's serum were noticeably higher than those of the healthy controls (P < 0.05). PPs provide the microenvironment to induce the production of IgA-secreting plasmablasts. CONCLUSION TFH cells may be a key moderator to induce B cell differentiation into IgAsecreting plasmablasts and produce Gd-IgA1, which plays a significant part in IgAN's pathogenesis. It could be a new therapeutic target in the future.
Collapse
Affiliation(s)
- Yuye Huang
- Department of Pediatrics, General Hospital of Central Theater Command, No. 627, Wuluo Road, Wuhan 430030, Hubei Province, P.R. China
| | - Xunling Sun
- Department of Pediatrics, General Hospital of Central Theater Command, No. 627, Wuluo Road, Wuhan 430030, Hubei Province, P.R. China
| | - Guoming Nie
- Department of Pediatrics, General Hospital of Central Theater Command, No. 627, Wuluo Road, Wuhan 430030, Hubei Province, P.R. China
| | - Hongtao Xu
- Department of Pediatrics, General Hospital of Central Theater Command, No. 627, Wuluo Road, Wuhan 430030, Hubei Province, P.R. China
| | - Minshu Zou
- Department of Pediatrics, General Hospital of Central Theater Command, No. 627, Wuluo Road, Wuhan 430030, Hubei Province, P.R. China
| |
Collapse
|
32
|
O'Neill A, Mantri CK, Tan CW, Saron WAA, Nagaraj SK, Kala MP, Joy CM, Rathore APS, Tripathi S, Wang LF, St John AL. Mucosal SARS-CoV-2 vaccination of rodents elicits superior systemic T central memory function and cross-neutralising antibodies against variants of concern. EBioMedicine 2024; 99:104924. [PMID: 38113758 PMCID: PMC10772395 DOI: 10.1016/j.ebiom.2023.104924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 11/27/2023] [Accepted: 12/02/2023] [Indexed: 12/21/2023] Open
Abstract
BACKGROUND COVID-19 vaccines used in humans are highly effective in limiting disease and death caused by the SARS-CoV-2 virus, yet improved vaccines that provide greater protection at mucosal surfaces, which could reduce break-through infections and subsequent transmission, are still needed. METHODS Here we tested an intranasal (I.N.) vaccination with the receptor binding domain of Spike antigen of SARS-CoV-2 (S-RBD) in combination with the mucosal adjuvant mastoparan-7 compared with the sub-cutaneous (S.C.) route, adjuvanted by either M7 or the gold-standard adjuvant, alum, in mice, for immunological read-outs. The same formulation delivered I.N. or S.C. was tested in hamsters to assess efficacy. FINDINGS I.N. vaccination improved systemic T cell responses compared to an equivalent dose of antigen delivered S.C. and T cell phenotypes induced by I.N. vaccine administration included enhanced polyfunctionality (combined IFN-γ and TNF expression) and greater numbers of T central memory (TCM) cells. These phenotypes were T cell-intrinsic and could be recalled in the lungs and/or brachial LNs upon antigen challenge after adoptive T cell transfer to naïve recipients. Furthermore, mucosal vaccination induced antibody responses that were similarly effective in neutralising the binding of the parental strain of S-RBD to its ACE2 receptor, but showed greater cross-neutralising capacity against multiple variants of concern (VOC), compared to S.C. vaccination. I.N. vaccination provided significant protection from lung pathology compared to unvaccinated animals upon challenge with homologous and heterologous SARS-CoV-2 strains in a hamster model. INTERPRETATION These results highlight the role of nasal vaccine administration in imprinting an immune profile associated with long-term T cell retention and diversified neutralising antibody responses, which could be applied to improve vaccines for COVID-19 and other infectious diseases. FUNDING This study was funded by Duke-NUS Medical School, the Singapore Ministry of Education, the National Medical Research Council of Singapore and a DBT-BIRAC Grant.
Collapse
Affiliation(s)
- Aled O'Neill
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, 169857, Singapore
| | - Chinmay Kumar Mantri
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, 169857, Singapore
| | - Chee Wah Tan
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, 169857, Singapore; Infectious Diseases Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 117545, Singapore
| | - Wilfried A A Saron
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, 169857, Singapore
| | - Santhosh Kambaiah Nagaraj
- Centre for Infectious Disease Research, Microbiology and Cell Biology Department, Indian Institute of Science, Bengaluru, 560012, India
| | - Monica Palanichamy Kala
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, 169857, Singapore
| | - Christy Margarat Joy
- Centre for Infectious Disease Research, Microbiology and Cell Biology Department, Indian Institute of Science, Bengaluru, 560012, India
| | - Abhay P S Rathore
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, 169857, Singapore; Department of Pathology, Duke University Medical Centre, Durham, North Carolina, 27705, USA
| | - Shashank Tripathi
- Centre for Infectious Disease Research, Microbiology and Cell Biology Department, Indian Institute of Science, Bengaluru, 560012, India
| | - Lin-Fa Wang
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, 169857, Singapore; SingHealth Duke-NUS Global Health Institute, Singapore
| | - Ashley L St John
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, 169857, Singapore; Department of Pathology, Duke University Medical Centre, Durham, North Carolina, 27705, USA; SingHealth Duke-NUS Global Health Institute, Singapore; Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
33
|
Xie Q, Liao X, Huang B, Wang L, Liao G, Luo C, Wen S, Fang S, Luo H, Shu Y. The truncated IFITM3 facilitates the humoral immune response in inactivated influenza vaccine-vaccinated mice via interaction with CD81. Emerg Microbes Infect 2023; 12:2246599. [PMID: 37556756 PMCID: PMC10484049 DOI: 10.1080/22221751.2023.2246599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 07/19/2023] [Accepted: 08/06/2023] [Indexed: 08/11/2023]
Abstract
A single-nucleotide polymorphism (SNP) rs12252-C of interferon-induced transmembrane protein 3 (IFITM3), resulting in a truncated IFITM3 protein lacking 21 N-terminus amino acids, is associated with severe influenza infection in the Chinese population. However, the effect of IFITM3 rs12252-C on influenza vaccination and the underlying mechanism is poorly understood. Here, we constructed a mouse model with a deletion of 21 amino acids at the N-terminus (NΔ21) of IFITM3 and then compared the antibody response between Quadrivalent influenza vaccine (QIV) immunized wild-type (WT) mice and NΔ21 mice. Significantly higher levels of haemagglutination inhibition (HI) titre, neutralizing antibodies (NAb), and immunoglobulin G (IgG) to H1N1, H3N2, B/Victory, and B/Yamagata viruses were observed in NΔ21 mice compared to WT mice. Correspondingly, the numbers of splenic germinal centre (GC) B cells, plasma cells, memory B cells, QIV-specific IgG+ antibody-secreting cells (ASC), and T follicular helper cells (TFH) in NΔ21 mice were higher compared with WT mice. Moreover, the 21-amino-acid deletion caused IFITM3 translocation from the endocytosis compartment to the periphery of cells, which also prevented the degradation of a co-stimulatory molecule of B cell receptor (BCR) CD81 on the cell surface. More importantly, a more interaction was observed between NΔ21 protein and CD81 compared to the interaction between IFITM3 and CD81. Overall, our study revealed a potential mechanism of NΔ21 protein enhancing humoral immune response by relocation to prevent the degradation of CD81, providing insight into SNP affecting influenza vaccination.
Collapse
Affiliation(s)
- Qian Xie
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, People’s Republic of China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Xinzhong Liao
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, People’s Republic of China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Bi Huang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, People’s Republic of China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Liangliang Wang
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, People’s Republic of China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Guancheng Liao
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, People’s Republic of China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Chuming Luo
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, People’s Republic of China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Simin Wen
- Guangzhou First People’s Hospital, the Second Affiliated Hospital of South China University of Technology, Guangzhou, People’s Republic of China
| | - Shisong Fang
- Pathogenic Microorganism Testing Institute, Shenzhen Center for Disease Control and Prevention, Shenzhen, People’s Republic of China
| | - Huanle Luo
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, People’s Republic of China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, People’s Republic of China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, People’s Republic of China
| | - Yuelong Shu
- School of Public Health (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, People’s Republic of China
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, People’s Republic of China
- Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
| |
Collapse
|
34
|
Shigehara K, Kamekura R, Ikegami I, Sakamoto H, Yanagi M, Kamiya S, Kodama K, Asai Y, Miyajima S, Nishikiori H, Uno E, Yamamoto K, Takano K, Chiba H, Ohnishi H, Ichimiya S. Circulating T follicular helper 2 cells, T follicular regulatory cells and regulatory B cells are effective biomarkers for predicting the response to house dust mite sublingual immunotherapy in patients with allergic respiratory diseases. Front Immunol 2023; 14:1284205. [PMID: 38111589 PMCID: PMC10726700 DOI: 10.3389/fimmu.2023.1284205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 11/06/2023] [Indexed: 12/20/2023] Open
Abstract
The relationships between T follicular helper (Tfh) cells and antigen-specific immunoglobulins (sIgs) in patients with allergic respiratory diseases who are receiving antigen immunotherapy (AIT) have not been fully clarified. Therefore, we started to perform house dust mite sublingual immunotherapy (HDM-SLIT) for 20 patients with atopic asthma comorbid with allergic rhinitis (AA+AR) who were already receiving ordinary treatments including inhaled corticosteroid (ICS). We examined percentages of circulating T follicular helper (cTfh) and regulatory (cTfr) cells and percentages of circulating regulatory T (cTreg) and B (cBreg) cells by FACS and we examined levels of Der-p/f sIgs by ELISA. Based on the symptom score (asthma control questionnaire: ACQ) and medication score ((global initiative for asthma: GINA) treatment step score) in patients with AA, the patients were divided into responders and non-responders. The percentage of cTfh2 cells significantly decreased and the percentage of cTfh1 cells significantly increased within the first year. Der-p/f sIgEs decreased after a transient elevation at 3 months in both groups. Notably, the percentage of cTfh2 cells and the ratio of cTfh2/cBreg cells and Der-p/f sIgEs greatly decreased in responders from 6 months to 12 months. The percentages of cTfr and cTreg cells showed significant negative correlations with the percentage of cTfh2 cells. The percentage of IL-4+ cTfh cells were significantly decreased and the percentage of IFN-γ+ cTfh cells were increased before treatment to 24 months in 6 patients examined (4 responders and 2 non-responders). We performed multi plelogistic regression analysis based on these results, the ratios of cTfh2/cTfr cells and cTfh2/cBreg cells at the start of therapy were statistically effective biomarkers for predicting the response to HDM-SLIT in patients with AA+AR.
Collapse
Affiliation(s)
- Katsunori Shigehara
- Department of Human Immunology, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, Sapporo, Japan
- Ai Medical Clinic, Sapporo, Japan
| | - Ryuta Kamekura
- Department of Human Immunology, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Otolaryngology and Head and Neck Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Ippei Ikegami
- Department of Human Immunology, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hiroshi Sakamoto
- Department of Human Immunology, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Otolaryngology and Head and Neck Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Masahiro Yanagi
- Department of Human Immunology, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Shiori Kamiya
- Department of Human Immunology, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
- Department of Dermatology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Kentaro Kodama
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yuichiro Asai
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Satsuki Miyajima
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hirotaka Nishikiori
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Eiji Uno
- Ai Medical Clinic, Sapporo, Japan
| | - Keisuke Yamamoto
- Department of Otolaryngology and Head and Neck Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Kenichi Takano
- Department of Otolaryngology and Head and Neck Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hirofumi Chiba
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hirofumi Ohnishi
- Department of Public Health, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Shingo Ichimiya
- Department of Human Immunology, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
35
|
Xu Y, Yan X, Wei T, Chen M, Zhu J, Gao J, Liu B, Zhu W, Liu Z. Transmucosal Delivery of Nasal Nanovaccines Enhancing Mucosal and Systemic Immunity. NANO LETTERS 2023; 23:10522-10531. [PMID: 37943583 DOI: 10.1021/acs.nanolett.3c03419] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2023]
Abstract
Intranasal vaccines can induce protective immune responses at the mucosa surface entrance, preventing the invasion of respiratory pathogens. However, the nasal barrier remains a major challenge in the development of intranasal vaccines. Herein, a transmucosal nanovaccine based on cationic fluorocarbon modified chitosan (FCS) is developed to induce mucosal immunity. In our system, FCS can self-assemble with the model antigen ovalbumin and TLR9 agonist CpG, effectively promoting the maturation and cross-presentation of dendritic cells. More importantly, it can enhance the production of secretory immunoglobin A (sIgA) at mucosal surfaces for those intranasally vaccinated mice, which in the meantime showed effective production of immunoglobulin G (IgG) systemically. As a proof-of-concept study, such a mucosal vaccine inhibits ovalbumin-expressing B16-OVA melanoma, especially its lung metastases. Our work presents a unique intranasal delivery system to deliver antigen across mucosal epithelia and promote mucosal and systemic immunity.
Collapse
Affiliation(s)
- Yuchun Xu
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Xiaoying Yan
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Ting Wei
- Suzhou InnoBM Pharmaceutics Co. Ltd., Suzhou, Jiangsu 215213, China
| | - Minming Chen
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Jiafei Zhu
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Juxin Gao
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Bo Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Wenjun Zhu
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Jiangsu Key Laboratory for Carbon-based Functional Materials and Devices, Soochow University, Suzhou 215123, China
- Suzhou InnoBM Pharmaceutics Co. Ltd., Suzhou, Jiangsu 215213, China
| |
Collapse
|
36
|
Merkenschlager J, Berz RM, Ramos V, Uhlig M, MacLean AJ, Nowosad CR, Oliveira TY, Nussenzweig MC. Continually recruited naïve T cells contribute to the follicular helper and regulatory T cell pools in germinal centers. Nat Commun 2023; 14:6944. [PMID: 37907454 PMCID: PMC10618265 DOI: 10.1038/s41467-023-41880-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 09/20/2023] [Indexed: 11/02/2023] Open
Abstract
Follicular helper T cells (TFH) mediate B cell selection and clonal expansion in germinal centers (GCs), and follicular regulatory T cells (TFR) prevent the emergence of self-reactive B cells and help to extinguish the reaction. Here we show that GC reactions continually recruit T cells from both the naïve conventional and naive thymic regulatory T cell (Treg) repertoires. In the early GC, newly recruited T cells develop into TFH, whereas cells entering during the contraction phase develop into TFR cells that contribute to GC dissolution. The TFR fate decision is associated with decreased antigen availability and is modulated by slow antigen delivery or mRNA vaccination. Thus, invasion of ongoing GCs by newly developing TFH and TFR helps remodel the GC based on antigen availability.
Collapse
Affiliation(s)
- Julia Merkenschlager
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, 10065, USA.
| | - Riza-Maria Berz
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, 10065, USA
| | - Victor Ramos
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, 10065, USA
| | - Maximilian Uhlig
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, 10065, USA
| | - Andrew J MacLean
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, 10065, USA
| | - Carla R Nowosad
- Translational Immunology Center, New York University Grossman School of Medicine, New York, NY, 10016, USA
| | - Thiago Y Oliveira
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, 10065, USA
| | - Michel C Nussenzweig
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, 10065, USA
- Howard Hughes Medical Institute, The Rockefeller University, New York, NY, 10065, USA
| |
Collapse
|
37
|
Xie L, Fang J, Yu J, Zhang W, He Z, Ye L, Wang H. The role of CD4 + T cells in tumor and chronic viral immune responses. MedComm (Beijing) 2023; 4:e390. [PMID: 37829505 PMCID: PMC10565399 DOI: 10.1002/mco2.390] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 09/06/2023] [Accepted: 09/12/2023] [Indexed: 10/14/2023] Open
Abstract
Immunotherapies are mainly aimed to promote a CD8+ T cell response rather than a CD4+ T cell response as cytotoxic T lymphocytes (CTLs) can directly kill target cells. Recently, CD4+ T cells have received more attention due to their diverse roles in tumors and chronic viral infections. In antitumor and antichronic viral responses, CD4+ T cells relay help signals through dendritic cells to indirectly regulate CD8+ T cell response, interact with B cells or macrophages to indirectly modulate humoral immunity or macrophage polarization, and inhibit tumor blood vessel formation. Additionally, CD4+ T cells can also exhibit direct cytotoxicity toward target cells. However, regulatory T cells exhibit immunosuppression and CD4+ T cells become exhausted, which promote tumor progression and chronic viral persistence. Finally, we also outline immunotherapies based on CD4+ T cells, including adoptive cell transfer, vaccines, and immune checkpoint blockade. Overall, this review summarizes diverse roles of CD4+ T cells in the antitumor or protumor and chronic viral responses, and also highlights the immunotherapies based on CD4+ T cells, giving a better understanding of their roles in tumors and chronic viral infections.
Collapse
Affiliation(s)
- Luoyingzi Xie
- Institute of Hepatopancreatobiliary SurgeryChongqing General HospitalChongqingChina
- The Institute of ImmunologyThird Military Medical University (Army Medical University)ChongqingChina
| | - Jingyi Fang
- The Institute of ImmunologyThird Military Medical University (Army Medical University)ChongqingChina
| | - Juncheng Yu
- Department of Thoracic SurgeryXinqiao Hospital Third Military Medical University (Army Medical University)ChongqingChina
| | - Weinan Zhang
- Department of Plastic & Cosmetic SurgeryArmy Medical Center of PLAAmy Medical UniversityChongqingChina
| | - Zhiqiang He
- Department of Plastic & Cosmetic SurgeryArmy Medical Center of PLAAmy Medical UniversityChongqingChina
| | - Lilin Ye
- The Institute of ImmunologyThird Military Medical University (Army Medical University)ChongqingChina
| | - Huaizhi Wang
- Institute of Hepatopancreatobiliary SurgeryChongqing General HospitalChongqingChina
| |
Collapse
|
38
|
Zeng S, Crichton ES, Ford ML, Badell IR. Memory T follicular helper cells drive donor-specific antibodies independent of memory B cells and primary germinal center and alloantibody formation. Am J Transplant 2023; 23:1511-1525. [PMID: 37302575 PMCID: PMC11228286 DOI: 10.1016/j.ajt.2023.06.006] [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: 04/14/2023] [Revised: 06/01/2023] [Accepted: 06/06/2023] [Indexed: 06/13/2023]
Abstract
Human leukocyte antigen antibodies are important immunologic mediators of renal allograft loss and are difficult to control. The inability to permanently eliminate donor-specific antibodies (DSA) is partly due to an incomplete understanding of the cellular mechanisms driving alloantibody formation, recurrence, and maintenance. Memory T follicular helper (mTfh) cells rapidly interact with memory B cells upon antigen re-exposure for anamnestic humoral responses, but little is known about Tfh memory in transplantation. We hypothesized that alloreactive mTfh cells form after transplantation and play a critical role in DSA formation following alloantigen re-encounter. To test this hypothesis, we utilized murine skin allograft models to identify and characterize Tfh memory and interrogate its ability to mediate alloantibody responses. We identified alloreactive Tfh memory as a mediator of accelerated humoral alloresponses independent of memory B cells and primary germinal center, or DSA, formation. Furthermore, we demonstrate that mTfh-driven alloantibody formation is susceptible to CD28 costimulation blockade. These findings provide novel insight into a pathologic role for memory Tfh in alloantibody responses and strongly support shifting therapeutic focus from the singular targeting of B cell lineage cells and alloantibodies themselves to multimodal strategies that include inhibition of mTfh cells to treat DSA.
Collapse
Affiliation(s)
- Shan Zeng
- Emory Transplant Center, Atlanta, Georgia, USA
| | | | | | | |
Collapse
|
39
|
Moya-Guzmán MJ, de Solminihac J, Padilla C, Rojas C, Pinto C, Himmel T, Pino-Lagos K. Extracellular Vesicles from Immune Cells: A Biomedical Perspective. Int J Mol Sci 2023; 24:13775. [PMID: 37762077 PMCID: PMC10531060 DOI: 10.3390/ijms241813775] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/04/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
Research on the role of extracellular vesicles (sEV) in physiology has demonstrated their undoubted importance in processes such as the transportation of molecules with significance for cell metabolism, cell communication, and the regulation of mechanisms such as cell differentiation, inflammation, and immunity. Although the role of EVs in the immune response is actively investigated, there is little literature revising, in a comprehensive manner, the role of small EVs produced by immune cells. Here, we present a review of studies reporting the release of sEV by different types of leukocytes and the implications of such observations on cellular homeostasis. We also discuss the function of immune cell-derived sEV and their relationship with pathological states, highlighting their potential application in the biomedical field.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Karina Pino-Lagos
- Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Av. Plaza 2501, Las Condes, Santiago 755000, Chile
| |
Collapse
|
40
|
Yadav M, Uikey BN, Rathore SS, Gupta P, Kashyap D, Kumar C, Shukla D, Vijayamahantesh, Chandel AS, Ahirwar B, Singh AK, Suman SS, Priyadarshi A, Amit A. Role of cytokine in malignant T-cell metabolism and subsequent alternation in T-cell tumor microenvironment. Front Oncol 2023; 13:1235711. [PMID: 37746258 PMCID: PMC10513393 DOI: 10.3389/fonc.2023.1235711] [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: 06/06/2023] [Accepted: 08/14/2023] [Indexed: 09/26/2023] Open
Abstract
T cells are an important component of adaptive immunity and T-cell-derived lymphomas are very complex due to many functional sub-types and functional elasticity of T-cells. As with other tumors, tissues specific factors are crucial in the development of T-cell lymphomas. In addition to neoplastic cells, T- cell lymphomas consist of a tumor micro-environment composed of normal cells and stroma. Numerous studies established the qualitative and quantitative differences between the tumor microenvironment and normal cell surroundings. Interaction between the various component of the tumor microenvironment is crucial since tumor cells can change the microenvironment and vice versa. In normal T-cell development, T-cells must respond to various stimulants deferentially and during these courses of adaptation. T-cells undergo various metabolic alterations. From the stage of quiescence to attention of fully active form T-cells undergoes various stage in terms of metabolic activity. Predominantly quiescent T-cells have ATP-generating metabolism while during the proliferative stage, their metabolism tilted towards the growth-promoting pathways. In addition to this, a functionally different subset of T-cells requires to activate the different metabolic pathways, and consequently, this regulation of the metabolic pathway control activation and function of T-cells. So, it is obvious that dynamic, and well-regulated metabolic pathways are important for the normal functioning of T-cells and their interaction with the microenvironment. There are various cell signaling mechanisms of metabolism are involved in this regulation and more and more studies have suggested the involvement of additional signaling in the development of the overall metabolic phenotype of T cells. These important signaling mediators include cytokines and hormones. The impact and role of these mediators especially the cytokines on the interplay between T-cell metabolism and the interaction of T-cells with their micro-environments in the context of T-cells lymphomas are discussed in this review article.
Collapse
Affiliation(s)
- Megha Yadav
- Department of Forensic Science, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | - Blessi N. Uikey
- Department of Forensic Science, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | | | - Priyanka Gupta
- Department of Forensic Science, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | - Diksha Kashyap
- Department of Forensic Science, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | - Chanchal Kumar
- Department of Forensic Science, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | - Dhananjay Shukla
- Department of Biotechnology, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | - Vijayamahantesh
- Department of Immunology and Microbiology, University of Missouri, Columbia, SC, United States
| | - Arvind Singh Chandel
- Center for Disease Biology and Integrative Medicine, Faculty of Medicine, The University of Tokyo, Bunkyo, Japan
| | - Bharti Ahirwar
- Department of Pharmacy, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | | | - Shashi Shekhar Suman
- Department of Zoology, Udayana Charya (UR) College, Lalit Narayan Mithila University, Darbhanga, India
| | - Amit Priyadarshi
- Department of Zoology, Veer Kunwar Singh University, Arrah, India
| | - Ajay Amit
- Department of Forensic Science, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| |
Collapse
|
41
|
Cui C, Craft J, Joshi NS. T follicular helper cells in cancer, tertiary lymphoid structures, and beyond. Semin Immunol 2023; 69:101797. [PMID: 37343412 DOI: 10.1016/j.smim.2023.101797] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 06/09/2023] [Accepted: 06/15/2023] [Indexed: 06/23/2023]
Abstract
With the emergence and success of checkpoint blockade immunotherapy, immuno-oncology has primarily focused on CD8 T cells, whose cytotoxic programs directly target tumor cells. However, the limited response rate of current immunotherapy regimens has prompted investigation into other types of tumor-infiltrating immune cells, such as CD4 T cells and B cells, and how they interact with CD8 T cells in a coordinated network. Recent studies have demonstrated the potential therapeutic benefits of CD4 T follicular helper (TFH) cells and B cells in cancer, highlighting the important role of their crosstalk and interactions with other immune cell components in the tumor microenvironment. These interactions also occur in tumor-associated tertiary lymphoid structures (TLS), which resemble secondary lymphoid organs (SLOs) with orchestrated vascular, chemokine, and cellular infrastructures that support the developmental pathways of functional immune cells. In this review, we discuss recent breakthroughs on TFH biology and T cell-B cell interactions in tumor immunology, and their potential as novel therapeutic targets to advance cancer treatment.
Collapse
Affiliation(s)
- Can Cui
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Joseph Craft
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Internal Medicine (Rheumatology, Allergy and Immunology), Yale University School of Medicine, New Haven, CT 06520, USA.
| | - Nikhil S Joshi
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
42
|
Zhou J, He X, Ou Y, Peng S, Li D, Zhou Q, Fu J, Long Y, Tan Y. Role of CXCR5 + CD8 + T cells in human hepatitis B virus infection. J Viral Hepat 2023; 30:638-645. [PMID: 37129474 DOI: 10.1111/jvh.13840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 04/05/2023] [Accepted: 04/15/2023] [Indexed: 05/03/2023]
Abstract
The replication of HBV in hepatocytes can be effectively inhibited by lifelong antiviral therapy. Because of the long-term presence of HBV reservoirs, the virus rebound frequently occurs once the treatment is stopped, which poses a considerable obstacle to the complete removal of the virus. In terms of gene composition, regulation of B cell action and function, CXCR5+ CD8+ T cells are similar to CXCR5+ CD4+ T follicular helper cells, while these cells are characterized by elevated programmed cell death 1 and cytotoxic-related proteins. CXCR5+ CD8+ T cells are strongly associated with progression in inflammatory and autoimmune diseases. In addition, CXCR5 expression on the surface of CD8+ T cells is mostly an indicator of memory stem cell-like failure in progenitor cells in cancer that are more responsive to immune checkpoint blocking therapy. Furthermore, the phenomena have also been demonstrated in some viral infections, highlighting the duality of the cellular immune response of CXCR5+ CD8+ T cells. This mini-review will focus on the function of CXCR5+ CD8+ T cells in HBV infection and discuss the function of these CD8+ T cells and the potential of associated co-stimulators or cytokines in HBV therapeutic strategies.
Collapse
Affiliation(s)
- Juan Zhou
- Department of Infectious Diseases, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, China
| | - Xiaojing He
- Department of Infectious Diseases, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, China
| | - Yangjing Ou
- Department of Infectious Diseases, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, China
| | - Shuang Peng
- Department of Infectious Diseases, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, China
| | - Dan Li
- Department of Infectious Diseases, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, China
| | - Qing Zhou
- Department of Infectious Diseases, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, China
| | - Jingli Fu
- Department of Infectious Diseases, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, China
| | - Yunzhu Long
- Department of Infectious Diseases, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, China
| | - Yingzheng Tan
- Department of Infectious Diseases, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, China
| |
Collapse
|
43
|
Zheng Z, Wieder T, Mauerer B, Schäfer L, Kesselring R, Braumüller H. T Cells in Colorectal Cancer: Unravelling the Function of Different T Cell Subsets in the Tumor Microenvironment. Int J Mol Sci 2023; 24:11673. [PMID: 37511431 PMCID: PMC10380781 DOI: 10.3390/ijms241411673] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/17/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Therapeutic options for metastatic colorectal cancer (mCRC) are very limited, and the prognosis using combination therapy with a chemotherapeutic drug and a targeted agent, e.g., epidermal growth factor receptor or tyrosine kinase, remains poor. Therefore, mCRC is associated with a poor median overall survival (mOS) of only 25-30 months. Current immunotherapies with checkpoint inhibitor blockade (ICB) have led to a substantial change in the treatment of several cancers, such as melanoma and non-small cell lung cancer. In CRC, ICB has only limited effects, except in patients with microsatellite instability-high (MSI-H) or mismatch repair-deficient (dMMR) tumors, which comprise about 15% of sporadic CRC patients and about 4% of patients with metastatic CRC. The vast majority of sporadic CRCs are microsatellite-stable (MSS) tumors with low levels of infiltrating immune cells, in which immunotherapy has no clinical benefit so far. Immunotherapy with checkpoint inhibitors requires the presence of infiltrating T cells into the tumor microenvironment (TME). This makes T cells the most important effector cells in the TME, as evidenced by the establishment of the immunoscore-a method to estimate the prognosis of CRC patients. The microenvironment of a tumor contains several types of T cells that are anti-tumorigenic, such as CD8+ T cells or pro-tumorigenic, such as regulatory T cells (Tregs) or T helper 17 (Th17) cells. However, even CD8+ T cells show marked heterogeneity, e.g., they can become exhausted, enter a state of hyporesponsiveness or become dysfunctional and express high levels of checkpoint molecules, the targets for ICB. To kill cancer cells, CD8+ T cells need the recognition of the MHC class I, which is often downregulated on colorectal cancer cells. In this case, a population of unconventional T cells with a γδ T cell receptor can overcome the limitations of the conventional CD8+ T cells with an αβT cell receptor. γδ T cells recognize antigens in an MHC-independent manner, thus acting as a bridge between innate and adaptive immunity. Here, we discuss the effects of different T cell subsets in colorectal cancer with a special emphasis on γδ T cells and the possibility of using them in CAR-T cell therapy. We explain T cell exclusion in microsatellite-stable colorectal cancer and the possibilities to overcome this exclusion to enable immunotherapy even in these "cold" tumors.
Collapse
Affiliation(s)
- Ziwen Zheng
- Department of General and Visceral Surgery, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Thomas Wieder
- Department of Vegetative and Clinical Physiology, Institute of Physiology, Eberhard Karls University Tübingen, 72074 Tübingen, Germany
| | - Bernhard Mauerer
- Department of General and Visceral Surgery, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- German Cancer Consortium (DKTK) Partner Site Freiburg, 79106 Freiburg, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Luisa Schäfer
- Department of General and Visceral Surgery, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Rebecca Kesselring
- Department of General and Visceral Surgery, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- German Cancer Consortium (DKTK) Partner Site Freiburg, 79106 Freiburg, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Heidi Braumüller
- Department of General and Visceral Surgery, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| |
Collapse
|
44
|
Stojanovic B, Jovanovic IP, Stojanovic MD, Jovanovic M, Vekic B, Milosevic B, Cvetkovic A, Spasic M, Stojanovic BS. The Emerging Roles of the Adaptive Immune Response in Acute Pancreatitis. Cells 2023; 12:1495. [PMID: 37296616 PMCID: PMC10253175 DOI: 10.3390/cells12111495] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 05/13/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
Acute pancreatitis (AP) is an abrupt, variable inflammatory condition of the pancreas, potentially escalating to severe systemic inflammation, rampant pancreatic necrosis, and multi-organ failure. Its complex pathogenesis involves an intricate immune response, with different T cell subsets (Th1, Th2, Th9, Th17, Th22, TFH, Treg, and CD8+ T cells) and B cells playing pivotal roles. Early T cell activation initiates the AP development, triggering cytokines associated with the Th1 response, which stimulate macrophages and neutrophils. Other T cell phenotypes contribute to AP's pathogenesis, and the balance between pro-inflammatory and anti-inflammatory cytokines influences its progression. Regulatory T and B cells are crucial for moderating the inflammatory response and promoting immune tolerance. B cells further contribute through antibody production, antigen presentation, and cytokine secretion. Understanding these immune cells' roles in AP could aid in developing new immunotherapies to enhance patient outcomes. However, further research is required to define these cells' precise roles in AP and their potential as therapeutic targets.
Collapse
Affiliation(s)
- Bojan Stojanovic
- Department of Surgery, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (B.S.)
| | - Ivan P. Jovanovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | | | - Marina Jovanovic
- Department of Internal Medicine, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Berislav Vekic
- Department of Surgery, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (B.S.)
| | - Bojan Milosevic
- Department of Surgery, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (B.S.)
| | - Aleksandar Cvetkovic
- Department of Surgery, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (B.S.)
| | - Marko Spasic
- Department of Surgery, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia; (B.S.)
| | - Bojana S. Stojanovic
- Department of Pathophysiology, Faculty of Medical Sciences, University of Kragujevac, 34000 Kragujevac, Serbia
| |
Collapse
|
45
|
Lin N, Yin W, Miller H, Byazrova MG, Herrada AA, Benlagha K, Lee P, Guan F, Lei J, Gong Q, Yan Y, Filatov A, Liu C. The role of regulatory T cells and follicular T helper cells in HBV infection. Front Immunol 2023; 14:1169601. [PMID: 37275865 PMCID: PMC10235474 DOI: 10.3389/fimmu.2023.1169601] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 04/20/2023] [Indexed: 06/07/2023] Open
Abstract
Hepatitis B has become one of the major global health threats, especially in developing countries and regions. Hepatitis B virus infection greatly increases the risk for liver diseases such as cirrhosis and cancer. However, treatment for hepatitis B is limited when considering the huge base of infected people. The immune response against hepatitis B is mediated mainly by CD8+ T cells, which are key to fighting invading viruses, while regulatory T cells prevent overreaction of the immune response process. Additionally, follicular T helper cells play a key role in B-cell activation, proliferation, differentiation, and formation of germinal centers. The pathogenic process of hepatitis B virus is generally the result of a disorder or dysfunction of the immune system. Therefore, we present in this review the critical functions and related biological processes of regulatory T cells and follicular T helper cells during HBV infection.
Collapse
Affiliation(s)
- Nengqi Lin
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Yin
- Wuhan Children’s Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Heather Miller
- Department of Research and Development, BD Biosciences, San Jose, CA, United States
| | - Maria G. Byazrova
- Laboratory of Immunochemistry, National Research Center Institute of Immunology, Federal Medical Biological Agency of Russia, Moscow, Russia
| | - Andrés A. Herrada
- Lymphatic Vasculature and Inflammation Research Laboratory, Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Talca, Chile
| | - Kamel Benlagha
- Université de Paris, Institut de Recherche Saint-Louis, EMiLy, Paris, France
| | - Pamela Lee
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Fei Guan
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Jiahui Lei
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| | - Quan Gong
- Department of Immunology, School of Medicine, Yangtze University, Jingzhou, China
- Clinical Molecular Immunology Center, School of Medicine, Yangtze University, Jingzhou, China
| | - Youqing Yan
- Department of Infectious Disease, Wuhan No.7 Hospital, Wuhan, China
| | - Alexander Filatov
- Laboratory of Immunochemistry, National Research Center Institute of Immunology, Federal Medical Biological Agency of Russia, Moscow, Russia
| | - Chaohong Liu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Disease, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
46
|
Seth A, Yokokura Y, Choi JY, Shyer JA, Vidyarthi A, Craft J. AP-1-independent NFAT signaling maintains follicular T cell function in infection and autoimmunity. J Exp Med 2023; 220:e20211110. [PMID: 36820828 PMCID: PMC9998660 DOI: 10.1084/jem.20211110] [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: 05/20/2021] [Revised: 09/05/2022] [Accepted: 02/01/2023] [Indexed: 02/24/2023] Open
Abstract
Coordinated gene expression programs enable development and function of T cell subsets. Follicular helper T (Tfh) cells coordinate humoral immune responses by providing selective and instructive cues to germinal center B cells. Here, we show that AP-1-independent NFAT gene expression, a program associated with hyporesponsive T cell states like anergy or exhaustion, is also a distinguishing feature of Tfh cells. NFAT signaling in Tfh cells, maintained by NFAT2 autoamplification, is required for their survival. ICOS signaling upregulates Bcl6 and induces an AP-1-independent NFAT program in primary T cells. Using lupus-prone mice, we demonstrate that genetic disruption or pharmacologic inhibition of NFAT signaling specifically impacts Tfh cell maintenance and leads to amelioration of autoantibody production and renal injury. Our data provide important conceptual and therapeutic insights into the signaling mechanisms that regulate Tfh cell development and function.
Collapse
Affiliation(s)
- Abhinav Seth
- Department of Internal Medicine, Section of Rheumatology, Allergy and Immunology, School of Medicine, Yale University, New Haven, CT, USA
| | - Yoshiyuki Yokokura
- Department of Internal Medicine, Section of Rheumatology, Allergy and Immunology, School of Medicine, Yale University, New Haven, CT, USA
| | - Jin-Young Choi
- Department of Internal Medicine, Section of Rheumatology, Allergy and Immunology, School of Medicine, Yale University, New Haven, CT, USA
| | - Justin A. Shyer
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
| | - Aurobind Vidyarthi
- Department of Internal Medicine, Section of Rheumatology, Allergy and Immunology, School of Medicine, Yale University, New Haven, CT, USA
| | - Joe Craft
- Department of Internal Medicine, Section of Rheumatology, Allergy and Immunology, School of Medicine, Yale University, New Haven, CT, USA
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
| |
Collapse
|
47
|
Yu M, Charles A, Cagigi A, Christ W, Österberg B, Falck-Jones S, Azizmohammadi L, Åhlberg E, Falck-Jones R, Svensson J, Nie M, Warnqvist A, Hellgren F, Lenart K, Arcoverde Cerveira R, Ols S, Lindgren G, Lin A, Maecker H, Bell M, Johansson N, Albert J, Sundling C, Czarnewski P, Klingström J, Färnert A, Loré K, Smed-Sörensen A. Delayed generation of functional virus-specific circulating T follicular helper cells correlates with severe COVID-19. Nat Commun 2023; 14:2164. [PMID: 37061513 PMCID: PMC10105364 DOI: 10.1038/s41467-023-37835-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 04/03/2023] [Indexed: 04/17/2023] Open
Abstract
Effective humoral immune responses require well-orchestrated B and T follicular helper (Tfh) cell interactions. Whether these interactions are impaired and associated with COVID-19 disease severity is unclear. Here, longitudinal blood samples across COVID-19 disease severity are analysed. We find that during acute infection SARS-CoV-2-specific circulating Tfh (cTfh) cells expand with disease severity. SARS-CoV-2-specific cTfh cell frequencies correlate with plasmablast frequencies and SARS-CoV-2 antibody titers, avidity and neutralization. Furthermore, cTfh cells but not other memory CD4 T cells, from severe patients better induce plasmablast differentiation and antibody production compared to cTfh cells from mild patients. However, virus-specific cTfh cell development is delayed in patients that display or later develop severe disease compared to those with mild disease, which correlates with delayed induction of high-avidity neutralizing antibodies. Our study suggests that impaired generation of functional virus-specific cTfh cells delays high-quality antibody production at an early stage, potentially enabling progression to severe disease.
Collapse
Affiliation(s)
- Meng Yu
- Division of Immunology and Allergy, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Afandi Charles
- Division of Immunology and Allergy, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Alberto Cagigi
- Division of Immunology and Allergy, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Wanda Christ
- Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Björn Österberg
- Division of Immunology and Allergy, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Sara Falck-Jones
- Division of Immunology and Allergy, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Lida Azizmohammadi
- Division of Immunology and Allergy, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Eric Åhlberg
- Division of Immunology and Allergy, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Ryan Falck-Jones
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Department of Perioperative Medicine and Intensive Care, Karolinska University Hospital, Stockholm, Sweden
| | - Julia Svensson
- Division of Immunology and Allergy, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Mu Nie
- Division of Immunology and Allergy, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Anna Warnqvist
- Division of Biostatistics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Fredrika Hellgren
- Division of Immunology and Allergy, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Klara Lenart
- Division of Immunology and Allergy, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Rodrigo Arcoverde Cerveira
- Division of Immunology and Allergy, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Sebastian Ols
- Division of Immunology and Allergy, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Gustaf Lindgren
- Division of Immunology and Allergy, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Ang Lin
- Division of Immunology and Allergy, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Holden Maecker
- The Human Immune Monitoring Center, Institute of Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - Max Bell
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Department of Perioperative Medicine and Intensive Care, Karolinska University Hospital, Stockholm, Sweden
| | - Niclas Johansson
- Division of Infectious Diseases, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Infectious Diseases, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Jan Albert
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
- Clinical Microbiology, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Christopher Sundling
- Division of Infectious Diseases, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Infectious Diseases, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Paulo Czarnewski
- Department of Biochemistry and Biophysics, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Stockholm University, Solna, Sweden
| | - Jonas Klingström
- Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Anna Färnert
- Division of Infectious Diseases, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Infectious Diseases, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Karin Loré
- Division of Immunology and Allergy, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Anna Smed-Sörensen
- Division of Immunology and Allergy, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
48
|
Rui K, Tang X, Shen Z, Jiang C, Zhu Q, Liu S, Che N, Tian J, Ling J, Yang Y. Exosome inspired photo-triggered gelation hydrogel composite on modulating immune pathogenesis for treating rheumatoid arthritis. J Nanobiotechnology 2023; 21:111. [PMID: 36973764 PMCID: PMC10044428 DOI: 10.1186/s12951-023-01865-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/20/2023] [Indexed: 03/29/2023] Open
Abstract
Although exosome therapy has been recognized as a promising strategy in the treatment of rheumatoid arthritis (RA), sustained modulation on RA specific pathogenesis and desirable protective effects for attenuating joint destruction still remain challenges. Here, silk fibroin hydrogel encapsulated with olfactory ecto-mesenchymal stem cell-derived exosomes (Exos@SFMA) was photo-crosslinked in situ to yield long-lasting therapeutic effect on modulating the immune microenvironment in RA. This in situ hydrogel system exhibited flexible mechanical properties and excellent biocompatibility for protecting tissue surfaces in joint. Moreover, the promising PD-L1 expression was identified on the exosomes, which potently suppressed Tfh cell polarization via inhibiting the PI3K/AKT pathway. Importantly, Exos@SFMA effectively relieved synovial inflammation and joint destruction by significantly reducing T follicular helper (Tfh) cell response and further suppressing the differentiation of germinal center (GC) B cells into plasma cells. Taken together, this exosome enhanced silk fibroin hydrogel provides an effective strategy for the treatment of RA and other autoimmune diseases.
Collapse
Affiliation(s)
- Ke Rui
- Institute of Medical Immunology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xiaoxuan Tang
- Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Ministry of Education and Jiangsu Province, Nantong University, Nantong, China
| | - Ziwei Shen
- Institute of Medical Immunology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Chao Jiang
- Institute of Medical Immunology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Qiugang Zhu
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Shiyi Liu
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Nan Che
- Department of Rheumatology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu, China
| | - Jie Tian
- Institute of Medical Immunology, Affiliated Hospital of Jiangsu University, Zhenjiang, China.
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China.
| | - Jue Ling
- Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Ministry of Education and Jiangsu Province, Nantong University, Nantong, China.
| | - Yumin Yang
- Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Ministry of Education and Jiangsu Province, Nantong University, Nantong, China.
| |
Collapse
|
49
|
Saeland E, van der Fits L, Bolder R, Heemskerk-van der Meer M, Drijver J, van Polanen Y, Vaneman C, Tettero L, Cox F, Serroyen J, Jorgensen MJ, Langedijk JPM, Schuitemaker H, Callendret B, Zahn RC. Combination Ad26.RSV.preF/preF protein vaccine induces superior protective immunity compared with individual vaccine components in preclinical models. NPJ Vaccines 2023; 8:45. [PMID: 36949051 PMCID: PMC10033289 DOI: 10.1038/s41541-023-00637-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 02/27/2023] [Indexed: 03/24/2023] Open
Abstract
Respiratory syncytial virus (RSV) is a leading cause of severe respiratory disease for which no licensed vaccine is available. We have previously shown that a prefusion (preF) conformation-stabilized RSV F protein antigen and an adenoviral vector encoding RSV preF protein (Ad26.RSV.preF) are immunogenic and protective in animals when administered as single components. Here, we evaluated a combination of the 2 components, administered as a single injection. Strong induction of both humoral and cellular responses was shown in RSV-naïve and pre-exposed mice and pre-exposed African green monkeys (AGMs). Both components of the combination vaccine contributed to humoral immune responses, while the Ad26.RSV.preF component was the main contributor to cellular immune responses in both mice and AGMs. Immunization with the combination elicited superior protection against RSV A2 challenge in cotton rats. These results demonstrate the advantage of a combination vaccine and support further clinical development.
Collapse
Affiliation(s)
- Eirikur Saeland
- Janssen Vaccines & Prevention B.V., Leiden, The Netherlands.
| | | | - Renske Bolder
- Janssen Vaccines & Prevention B.V., Leiden, The Netherlands
| | | | - Joke Drijver
- Janssen Vaccines & Prevention B.V., Leiden, The Netherlands
| | | | | | | | - Freek Cox
- Janssen Vaccines & Prevention B.V., Leiden, The Netherlands
| | - Jan Serroyen
- Janssen Vaccines & Prevention B.V., Leiden, The Netherlands
| | - Matthew J Jorgensen
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | | | | | | | - Roland C Zahn
- Janssen Vaccines & Prevention B.V., Leiden, The Netherlands
| |
Collapse
|
50
|
Liu T, Zheng M, Jia L, Wang M, Tang L, Wen Z, Zhang M, Yuan F. Deficient leptin receptor signaling in T cells of human SLE. Front Immunol 2023; 14:1157731. [PMID: 37006245 PMCID: PMC10063787 DOI: 10.3389/fimmu.2023.1157731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 03/06/2023] [Indexed: 03/19/2023] Open
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) is a prototypic autoimmune disease mainly mediated by IgG autoantibody. While follicular helper T (Tfh) cells are crucial for supporting IgG autoantibody generation in human SLE, underlying mechanisms for Tfh cell mal-differentiation remain unclear. METHODS In total, 129 SLE patients and 37 healthy donors were recruited for this study. Circulating leptin was determined by ELISA from patients with SLE and healthy individuals. CD4 T cells isolated from SLE patients and healthy donors were activated with anti-CD3/CD28 beads under cytokine-unbiased conditions in the presence or absence of recombinant leptin protein, followed by detection for Tfh cell differentiation by quantifying intracellular transcription factor Bcl-6 and cytokine IL-21. AMPK activation was assessed by analyzing phosphor-AMPK using phosflow cytometry and immunoblots. Leptin receptor expression was determined using flow cytometry and its overexpression was achieved by transfection with an expression vector. Humanized SLE chimeras were induced by injecting patients' immune cells into immune-deficient NSG mice and used for translational studies. RESULTS Circulating leptin was elevated in patients with SLE, inversely associated with disease activity. In healthy individuals, leptin efficiently inhibited Tfh cell differentiation through inducing AMPK activation. Meanwhile, leptin receptor deficiency was a feature of CD4 T cells in SLE patients, impairing the inhibitory effect of leptin on the differentiation of Tfh cells. As a result, we observed the coexistence of high circulating leptin and increased Tfh cell frequencies in SLE patients. Accordingly, overexpression of leptin receptor in SLE CD4 T cells abrogated Tfh cell mal-differentiation and IgG anti-dsDNA generation in humanized lupus chimeras. CONCLUSION Leptin receptor deficiency blocks the inhibitory effect of leptin on SLE Tfh cell differentiation, serving as a promising therapeutic target for lupus management.
Collapse
Affiliation(s)
- Ting Liu
- Department of Rheumatology, the Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
- Department of Rheumatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ming Zheng
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Li Jia
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Mingyuan Wang
- Department of Research Center, Suzhou Blood Center, Suzhou, China
| | - Longhai Tang
- Department of Research Center, Suzhou Blood Center, Suzhou, China
| | - Zhenke Wen
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou, China
| | - Miaojia Zhang
- Department of Rheumatology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Fenghong Yuan
- Department of Rheumatology, the Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| |
Collapse
|