1
|
Diamantopoulos N, Li J, Bouchard A, Joumier L, Mohammaei S, Panneton V, Chang J, Malleshaiah M, Suh WK. ICOS-expressing Regulatory T Cells Influence the Composition of Antitumor CTL Populations. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:753-762. [PMID: 38995175 DOI: 10.4049/jimmunol.2300154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 06/25/2024] [Indexed: 07/13/2024]
Abstract
The role of ICOS in antitumor T cell responses and overall tumor progression has been controversial. In this study, we compared tumor progression in mice lacking ICOS selectively in regulatory T (Treg) cells or in all T cells. Using an experimental melanoma lung metastasis model, we found that Treg cell-specific ICOS knockout reduces the overall tumor burden compared with Cre control mice, with increased CD4+-to-Treg cell and CD8+-to-Treg cell ratios in the tumor. In contrast, there was no difference in the tumor burden in mice lacking ICOS in all of the T cell compartments. This suggests a dual role of ICOS costimulation in promoting protumor and antitumor T cell responses. Consistent with reduced tumor burden, we found that Treg cell-specific deletion of ICOS leads to an increase of CD8+ CTLs that express high levels of granzyme B and perforin. Moreover, single-cell transcriptome analysis revealed an increase of Ly108+Eomeshi CD8+ T cells at the cost of the Ly108+T-bethi subset in Treg cell-specific knockout mice. These results suggest that ICOS-expressing Treg cells suppress the CTL maturation process at the level of Eomes upregulation, a critical step known to drive perforin expression and cytotoxicity. Collectively, our data imply that cancer immunotherapies using ICOS agonist Abs may work better in Treg cell-low tumors or when they are combined with regimens that deplete tumor-infiltrating Treg cells.
Collapse
Affiliation(s)
- Nikoletta Diamantopoulos
- Institut de Recherches Cliniques de Montréal, Montreal, Quebec, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
| | - Joanna Li
- Institut de Recherches Cliniques de Montréal, Montreal, Quebec, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
| | - Antoine Bouchard
- Institut de Recherches Cliniques de Montréal, Montreal, Quebec, Canada
- Molecular Biology Program, University of Montreal, Montreal, Quebec, Canada
| | - Loick Joumier
- Institut de Recherches Cliniques de Montréal, Montreal, Quebec, Canada
- Department of Biochemistry and Molecular Medicine, University of Montreal, Montreal, Quebec, Canada
| | - Saba Mohammaei
- Institut de Recherches Cliniques de Montréal, Montreal, Quebec, Canada
- Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada
| | - Vincent Panneton
- Institut de Recherches Cliniques de Montréal, Montreal, Quebec, Canada
- Department of Microbiology, Infectiology and Immunology, University of Montreal, Montreal, Quebec, Canada
| | - Jinsam Chang
- Institut de Recherches Cliniques de Montréal, Montreal, Quebec, Canada
- Molecular Biology Program, University of Montreal, Montreal, Quebec, Canada
| | - Mohan Malleshaiah
- Institut de Recherches Cliniques de Montréal, Montreal, Quebec, Canada
- Molecular Biology Program, University of Montreal, Montreal, Quebec, Canada
- Department of Biochemistry and Molecular Medicine, University of Montreal, Montreal, Quebec, Canada
- Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada
| | - Woong-Kyung Suh
- Institut de Recherches Cliniques de Montréal, Montreal, Quebec, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
- Molecular Biology Program, University of Montreal, Montreal, Quebec, Canada
- Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada
- Department of Microbiology, Infectiology and Immunology, University of Montreal, Montreal, Quebec, Canada
| |
Collapse
|
2
|
Ding Q, Xu Q, Hong Y, Zhou H, He X, Niu C, Tian Z, Li H, Zeng P, Liu J. Integrated analysis of single-cell RNA-seq, bulk RNA-seq, Mendelian randomization, and eQTL reveals T cell-related nomogram model and subtype classification in rheumatoid arthritis. Front Immunol 2024; 15:1399856. [PMID: 38962008 PMCID: PMC11219584 DOI: 10.3389/fimmu.2024.1399856] [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: 03/12/2024] [Accepted: 06/03/2024] [Indexed: 07/05/2024] Open
Abstract
Objective Rheumatoid arthritis (RA) is a systemic disease that attacks the joints and causes a heavy economic burden on humans worldwide. T cells regulate RA progression and are considered crucial targets for therapy. Therefore, we aimed to integrate multiple datasets to explore the mechanisms of RA. Moreover, we established a T cell-related diagnostic model to provide a new method for RA immunotherapy. Methods scRNA-seq and bulk-seq datasets for RA were obtained from the Gene Expression Omnibus (GEO) database. Various methods were used to analyze and characterize the T cell heterogeneity of RA. Using Mendelian randomization (MR) and expression quantitative trait loci (eQTL), we screened for potential pathogenic T cell marker genes in RA. Subsequently, we selected an optimal machine learning approach by comparing the nine types of machine learning in predicting RA to identify T cell-related diagnostic features to construct a nomogram model. Patients with RA were divided into different T cell-related clusters using the consensus clustering method. Finally, we performed immune cell infiltration and clinical correlation analyses of T cell-related diagnostic features. Results By analyzing the scRNA-seq dataset, we obtained 10,211 cells that were annotated into 7 different subtypes based on specific marker genes. By integrating the eQTL from blood and RA GWAS, combined with XGB machine learning, we identified a total of 8 T cell-related diagnostic features (MIER1, PPP1CB, ICOS, GADD45A, CD3D, SLFN5, PIP4K2A, and IL6ST). Consensus clustering analysis showed that RA could be classified into two different T-cell patterns (Cluster 1 and Cluster 2), with Cluster 2 having a higher T-cell score than Cluster 1. The two clusters involved different pathways and had different immune cell infiltration states. There was no difference in age or sex between the two different T cell patterns. In addition, ICOS and IL6ST were negatively correlated with age in RA patients. Conclusion Our findings elucidate the heterogeneity of T cells in RA and the communication role of these cells in an RA immune microenvironment. The construction of T cell-related diagnostic models provides a resource for guiding RA immunotherapeutic strategies.
Collapse
Affiliation(s)
- Qiang Ding
- The First School of Clinical Medicine, Guangxi Traditional Chinesen Medical University, Nanning, China
| | - Qingyuan Xu
- The First School of Clinical Medicine, Guangxi Traditional Chinesen Medical University, Nanning, China
| | - Yini Hong
- Gynecology Department, The First People’s Hospital of Guangzhou, Guangzhou, China
| | - Honghai Zhou
- Faculty of Orthopedics and Traumatology, Guangxi University of Chinese Medicine, Nanning, China
| | - Xinyu He
- The First School of Clinical Medicine, Guangxi Traditional Chinesen Medical University, Nanning, China
| | - Chicheng Niu
- The First School of Clinical Medicine, Guangxi Traditional Chinesen Medical University, Nanning, China
| | - Zhao Tian
- The First School of Clinical Medicine, Guangxi Traditional Chinesen Medical University, Nanning, China
| | - Hao Li
- The First School of Clinical Medicine, Guangxi Traditional Chinesen Medical University, Nanning, China
| | - Ping Zeng
- Department of Orthopedics and Traumatology, The First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, Guangxi, China
| | - Jinfu Liu
- Department of Orthopedics and Traumatology, The First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, Guangxi, China
| |
Collapse
|
3
|
Lu J, Zhou H, Chen Y, Xia X, Yang J, Ma J, Tian J, Wang S. Tfh cell-derived small extracellular vesicles exacerbate the severity of collagen-induced arthritis by enhancing B-cell responses. J Autoimmun 2024; 146:103235. [PMID: 38696926 DOI: 10.1016/j.jaut.2024.103235] [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: 11/23/2023] [Revised: 03/28/2024] [Accepted: 04/16/2024] [Indexed: 05/04/2024]
Abstract
Soluble components secreted by Tfh cells are critical for the germinal center responses. In this study, we investigated whether Tfh cells could regulate the B-cell response by releasing small extracellular vesicles (sEVs). Our results showed that Tfh cells promote B-cell differentiation and antibody production through sEVs and that CD40L plays a crucial role in Tfh-sEVs function. In addition, increased Tfh-sEVs were found in mice with collagen-induced arthritis (CIA). Adoptive transfer of Tfh cells significantly exacerbated the severity of CIA; however, the effect of Tfh cells on exacerbating the CIA process was significantly diminished after inhibiting sEVs secretion. Moreover, the levels of plasma Tfh-like-sEVs and CD40L expression on Tfh-like-sEVs in RA patients were significantly higher than those in healthy subjects. In summary, Tfh cell-derived sEVs can enhance the B-cell response, and exacerbate the procession of autoimmune arthritis.
Collapse
Affiliation(s)
- Jian Lu
- Department of Laboratory Medicine, Affiliated Hospital, Jiangsu University, Zhenjiang, China; Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Huimin Zhou
- Department of Laboratory Medicine, Affiliated Hospital, Jiangsu University, Zhenjiang, China; Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yuxuan Chen
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Xueli Xia
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Jun Yang
- Department of Laboratory Medicine, Affiliated People's Hospital, Jiangsu University, Zhenjiang, China
| | - Jie Ma
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Jie Tian
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China.
| | - Shengjun Wang
- Department of Laboratory Medicine, Affiliated Hospital, Jiangsu University, Zhenjiang, China; Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China; Department of Laboratory Medicine, Affiliated People's Hospital, Jiangsu University, Zhenjiang, China.
| |
Collapse
|
4
|
Yang Q, Zhang F, Chen H, Hu Y, Yang N, Yang W, Wang J, Yang Y, Xu R, Xu C. The differentiation courses of the Tfh cells: a new perspective on autoimmune disease pathogenesis and treatment. Biosci Rep 2024; 44:BSR20231723. [PMID: 38051200 PMCID: PMC10830446 DOI: 10.1042/bsr20231723] [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: 10/06/2023] [Revised: 11/23/2023] [Accepted: 12/04/2023] [Indexed: 12/07/2023] Open
Abstract
The follicular helper T cells are derived from CD4+T cells, promoting the formation of germinal centers and assisting B cells to produce antibodies. This review describes the differentiation process of Tfh cells from the perspectives of the initiation, maturation, migration, efficacy, and subset classification of Tfh cells, and correlates it with autoimmune disease, to provide information for researchers to fully understand Tfh cells and provide further research ideas to manage immune-related diseases.
Collapse
Affiliation(s)
- Qingya Yang
- Division of Rheumatology, People’s Hospital of Mianzhu, Mianzhu, Sichuan, 618200, China
| | - Fang Zhang
- Division of Rheumatology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210028, China
- Division of Rheumatology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, Jiangsu 210028, China
| | - Hongyi Chen
- Division of Rheumatology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210028, China
- Division of Rheumatology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, Jiangsu 210028, China
| | - Yuman Hu
- Division of Rheumatology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210028, China
- Division of Rheumatology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, Jiangsu 210028, China
| | - Ning Yang
- Division of Rheumatology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210028, China
- Division of Rheumatology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, Jiangsu 210028, China
| | - Wenyan Yang
- Division of Rheumatology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210028, China
- Division of Rheumatology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, Jiangsu 210028, China
| | - Jing Wang
- Division of Rheumatology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210028, China
- Division of Rheumatology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, Jiangsu 210028, China
| | - Yaxu Yang
- Division of Rheumatology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210028, China
- Division of Rheumatology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, Jiangsu 210028, China
| | - Ran Xu
- Division of Rheumatology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210028, China
- Division of Rheumatology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, Jiangsu 210028, China
| | - Chao Xu
- Division of Rheumatology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210028, China
- Division of Rheumatology, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, Jiangsu 210028, China
| |
Collapse
|
5
|
Small A, Lowe K, Wechalekar MD. Immune checkpoints in rheumatoid arthritis: progress and promise. Front Immunol 2023; 14:1285554. [PMID: 38077329 PMCID: PMC10704353 DOI: 10.3389/fimmu.2023.1285554] [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: 08/30/2023] [Accepted: 11/07/2023] [Indexed: 12/18/2023] Open
Abstract
Rheumatoid arthritis (RA) is one of the most prevalent autoimmune inflammatory conditions, and while the mechanisms driving pathogenesis are yet to be completely elucidated, self-reactive T cells and immune checkpoint pathways have a clear role. In this review, we provide an overview of the importance of checkpoint pathways in the T cell response and describe the involvement of these in RA development and progression. We discuss the relationship between immune checkpoint therapy in cancer and autoimmune adverse events, draw parallels with the involvement of immune checkpoints in RA pathobiology, summarise emerging research into some of the lesser-known pathways, and the potential of targeting checkpoint-related pathways in future treatment approaches to RA management.
Collapse
Affiliation(s)
- Annabelle Small
- Department of Rheumatology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Katie Lowe
- Department of Rheumatology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Mihir D Wechalekar
- Department of Rheumatology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
- Department of Rheumatology, Flinders Medical Centre, Adelaide, SA, Australia
| |
Collapse
|
6
|
Panneton V, Mindt BC, Bouklouch Y, Bouchard A, Mohammaei S, Chang J, Diamantopoulos N, Witalis M, Li J, Stancescu A, Bradley JE, Randall TD, Fritz JH, Suh WK. ICOS costimulation is indispensable for the differentiation of T follicular regulatory cells. Life Sci Alliance 2023; 6:e202201615. [PMID: 36754569 PMCID: PMC9909462 DOI: 10.26508/lsa.202201615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 01/24/2023] [Accepted: 01/25/2023] [Indexed: 02/10/2023] Open
Abstract
ICOS is a T-cell costimulatory receptor critical for Tfh cell generation and function. However, the role of ICOS in Tfr cell differentiation remains unclear. Using Foxp3-Cre-mediated ICOS knockout (ICOS FC) mice, we show that ICOS deficiency in Treg-lineage cells drastically reduces the number of Tfr cells during GC reactions but has a minimal impact on conventional Treg cells. Single-cell transcriptome analysis of Foxp3+ cells at an early stage of the GC reaction suggests that ICOS normally inhibits Klf2 expression to promote follicular features including Bcl6 up-regulation. Furthermore, ICOS costimulation promotes nuclear localization of NFAT2, a known driver of CXCR5 expression. Notably, ICOS FC mice had an unaltered overall GC B-cell output but showed signs of expanded autoreactive B cells along with elevated autoantibody titers. Thus, our study demonstrates that ICOS costimulation is critical for Tfr cell differentiation and highlights the importance of Tfr cells in maintaining humoral immune tolerance during GC reactions.
Collapse
Affiliation(s)
- Vincent Panneton
- Institut de Recherches Cliniques de Montréal, Quebec, Canada
- Department of Microbiology, Infectiology and Immunology, University of Montreal, Quebec, Canada
| | - Barbara C Mindt
- Department of Microbiology and Immunology, McGill University, Quebec, Canada
- McGill University Research Centre on Complex Traits, McGill University, Quebec, Canada
| | | | - Antoine Bouchard
- Institut de Recherches Cliniques de Montréal, Quebec, Canada
- Molecular Biology Program, University of Montreal, Quebec, Canada
| | - Saba Mohammaei
- Institut de Recherches Cliniques de Montréal, Quebec, Canada
- Division of Experimental Medicine, McGill University, Quebec, Canada
| | - Jinsam Chang
- Institut de Recherches Cliniques de Montréal, Quebec, Canada
- Molecular Biology Program, University of Montreal, Quebec, Canada
| | - Nikoletta Diamantopoulos
- Institut de Recherches Cliniques de Montréal, Quebec, Canada
- Department of Microbiology and Immunology, McGill University, Quebec, Canada
| | - Mariko Witalis
- Institut de Recherches Cliniques de Montréal, Quebec, Canada
- Molecular Biology Program, University of Montreal, Quebec, Canada
| | - Joanna Li
- Institut de Recherches Cliniques de Montréal, Quebec, Canada
- Department of Microbiology and Immunology, McGill University, Quebec, Canada
| | | | - John E Bradley
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Troy D Randall
- Department of Medicine, Division of Clinical Immunology and Rheumatology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jörg H Fritz
- Department of Microbiology and Immunology, McGill University, Quebec, Canada
- McGill University Research Centre on Complex Traits, McGill University, Quebec, Canada
| | - Woong-Kyung Suh
- Institut de Recherches Cliniques de Montréal, Quebec, Canada
- Department of Microbiology, Infectiology and Immunology, University of Montreal, Quebec, Canada
- Department of Microbiology and Immunology, McGill University, Quebec, Canada
- Molecular Biology Program, University of Montreal, Quebec, Canada
- Division of Experimental Medicine, McGill University, Quebec, Canada
| |
Collapse
|
7
|
Ding S, Sun Z, Jiang J, Chang X, Shen Y, Gu Y, Liu C. Inducible costimulator ligand (ICOSL) on CD19+ B cells is involved in immunopathological damage of rheumatoid arthritis (RA). Front Immunol 2022; 13:1015831. [DOI: 10.3389/fimmu.2022.1015831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 10/10/2022] [Indexed: 11/05/2022] Open
Abstract
Inducible costimulator (ICOS) and its ligand (ICOSL) are critical to regulate the immune response in autoimmune diseases. The participation of B lymphocytes exhibits pathogenic potential in the disease process of rheumatoid arthritis (RA). However, the precise role of ICOSL in RA remains unclear. In this study, we aimed to explore the regulatory effects of CD19+ICOSL+ B cells in the pathogenesis of RA. We demonstrated the increased expression of ICOS and ICOSL in patients with RA and collagen-induced arthritis (CIA) mice. The population of CD19+ICOSL+ B-cell subset was significantly correlated with clinicopathological characteristics of RA patients and CIA mice. Adoptive transfer of CD19+ICOSL+ B cells aggravated arthritic progression in CIA mice. Moreover, microarray analysis revealed that CD19+ICOSL+ cells could exert pivotal effect in pathological process of RA. Further blocking of ICOSL significantly inhibited proinflammatory responses and ameliorated arthritic progression. Therefore, CD19+ICOSL+ B-cell subset could be defined as a specific pathogenic cell subpopulation involved in immunopathological damage of RA. Blockade of ICOSL is promising to be a potential new approach for RA therapy.
Collapse
|
8
|
Chang J, Bouchard A, Bouklouch Y, Panneton V, Li J, Diamantopoulos N, Mohammaei S, Istomine R, Alvarez F, Piccirillo CA, Suh WK. ICOS-Deficient Regulatory T Cells Can Prevent Spontaneous Autoimmunity but Are Impaired in Controlling Acute Inflammation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:301-309. [PMID: 35760518 DOI: 10.4049/jimmunol.2100897] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 05/02/2022] [Indexed: 12/12/2022]
Abstract
ICOS is induced in activated T cells and its main role is to boost differentiation and function of effector T cells. ICOS is also constitutively expressed in a subpopulation of Foxp3+ regulatory T cells under steady-state condition. Studies using ICOS germline knockout mice or ICOS-blocking reagents suggested that ICOS has supportive roles in regulatory T (Treg) cell homeostasis, migration, and function. To avoid any compounding effects that may arise from ICOS-deficient non-Treg cells, we generated a conditional knockout system in which ICOS expression is selectively abrogated in Foxp3-expressing cells (ICOS FC mice). Compared to Foxp3-Cre control mice, ICOS FC mice showed a minor numerical deficit of steady-state Treg cells but did not show any signs of spontaneous autoimmunity, indicating that tissue-protective Treg populations do not heavily rely on ICOS costimulation. However, ICOS FC mice showed more severe inflammation in oxazolone-induced contact hypersensitivity, a model of atopic dermatitis. This correlated with elevated numbers of inflammatory T cells expressing IFN-γ and/or TNF-α in ICOS FC mice compared with the control group. In contrast, elimination of ICOS in all T cell compartments negated the differences, confirming that ICOS has a dual positive role in effector and Treg cells. Single-cell transcriptome analysis suggested that ICOS-deficient Treg cells fail to mature into T-bet+CXCR3+ "Th1-Treg" cells in the draining lymph node. Our results suggest that regimens that preferentially stimulate ICOS pathways in Treg cells might be beneficial for the treatment of Th1-driven inflammation.
Collapse
Affiliation(s)
- Jinsam Chang
- Institut de Recherches Cliniques de Montréal, Montreal, QC, Canada.,Molecular Biology Program, University of Montreal, Montreal, Quebec, Canada
| | - Antoine Bouchard
- Institut de Recherches Cliniques de Montréal, Montreal, QC, Canada.,Molecular Biology Program, University of Montreal, Montreal, Quebec, Canada
| | - Yasser Bouklouch
- Institut de Recherches Cliniques de Montréal, Montreal, QC, Canada
| | - Vincent Panneton
- Institut de Recherches Cliniques de Montréal, Montreal, QC, Canada.,Department of Microbiology, Infectiology and Immunology, University of Montreal, Montreal, Quebec, Canada
| | - Joanna Li
- Institut de Recherches Cliniques de Montréal, Montreal, QC, Canada.,Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada; and
| | - Nikoletta Diamantopoulos
- Institut de Recherches Cliniques de Montréal, Montreal, QC, Canada.,Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada; and
| | - Saba Mohammaei
- Institut de Recherches Cliniques de Montréal, Montreal, QC, Canada.,Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada
| | - Roman Istomine
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada; and
| | - Fernando Alvarez
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada; and
| | - Ciriaco A Piccirillo
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada; and
| | - Woong-Kyung Suh
- Institut de Recherches Cliniques de Montréal, Montreal, QC, Canada; .,Molecular Biology Program, University of Montreal, Montreal, Quebec, Canada.,Department of Microbiology, Infectiology and Immunology, University of Montreal, Montreal, Quebec, Canada.,Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada; and.,Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
9
|
Mayberry CL, Logan NA, Wilson JJ, Chang CH. Providing a Helping Hand: Metabolic Regulation of T Follicular Helper Cells and Their Association With Disease. Front Immunol 2022; 13:864949. [PMID: 35493515 PMCID: PMC9047778 DOI: 10.3389/fimmu.2022.864949] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 03/25/2022] [Indexed: 01/02/2023] Open
Abstract
T follicular helper (Tfh) cells provide support to B cells upon arrival in the germinal center, and thus are critical for the generation of a robust adaptive immune response. Tfh express specific transcription factors and cellular receptors including Bcl6, CXCR5, PD-1, and ICOS, which are critical for homing and overall function. Generally, the induction of an immune response is tightly regulated. However, deviation during this process can result in harmful autoimmunity or the inability to successfully clear pathogens. Recently, it has been shown that Tfh differentiation, activation, and proliferation may be linked with the cellular metabolic state. In this review we will highlight recent discoveries in Tfh differentiation and explore how these cells contribute to functional immunity in disease, including autoimmune-related disorders, cancer, and of particular emphasis, during infection.
Collapse
Affiliation(s)
| | | | | | - Chih-Hao Chang
- The Jackson Laboratory, Bar Harbor, ME, United States
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME, United States
- Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, MA, United States
- *Correspondence: Chih-Hao Chang,
| |
Collapse
|
10
|
Zhang X, Hu X, Tian T, Pang W. The role of ICOS in allergic disease: Positive or Negative? Int Immunopharmacol 2021; 103:108394. [PMID: 34922247 DOI: 10.1016/j.intimp.2021.108394] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/15/2021] [Accepted: 11/19/2021] [Indexed: 01/09/2023]
Abstract
With the rapid increase in the incidence of allergic diseases, the mechanisms underlying the development of these diseases have received a great deal of attention, and this is particularly true in regard to the role of ICOS in allergic diseases. Current studies have revealed that ICOS affects the functional activity of multiple immune cells that modulate the adaptive immune system. Additionally, ICOS also plays a crucial role in mediating cellular immunity and coordinating the response of the entire immune system, and thus, it plays a role in allergic reactions. However, the ICOS/ICOS-ligand (ICOS-L) axis functions in a dual role during the development of multiple allergic diseases. In this review, we explore the role of ICOS/ICOSL in the context of different immune cells that function in allergic diseases, and we summarize recent advances in their contribution to these diseases.
Collapse
Affiliation(s)
- Xueyan Zhang
- Department of Otorhinolaryngology-Head and Neck Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xianyang Hu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Tengfei Tian
- Department of Otolaryngology-Head and Neck Surgery, Eye Ear Nose and Throat Hospital, Fudan University, Shanghai, China
| | - Wenhui Pang
- Department of Otorhinolaryngology-Head and Neck Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
11
|
Zuo J, Tang J, Lu M, Zhou Z, Li Y, Tian H, Liu E, Gao B, Liu T, Shao P. Glycolysis Rate-Limiting Enzymes: Novel Potential Regulators of Rheumatoid Arthritis Pathogenesis. Front Immunol 2021; 12:779787. [PMID: 34899740 PMCID: PMC8651870 DOI: 10.3389/fimmu.2021.779787] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 11/02/2021] [Indexed: 01/10/2023] Open
Abstract
Rheumatoid arthritis (RA) is a classic autoimmune disease characterized by uncontrolled synovial proliferation, pannus formation, cartilage injury, and bone destruction. The specific pathogenesis of RA, a chronic inflammatory disease, remains unclear. However, both key glycolysis rate-limiting enzymes, hexokinase-II (HK-II), phosphofructokinase-1 (PFK-1), and pyruvate kinase M2 (PKM2), as well as indirect rate-limiting enzymes, 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 3 (PFKFB3), are thought to participate in the pathogenesis of RA. In here, we review the latest literature on the pathogenesis of RA, introduce the pathophysiological characteristics of HK-II, PFK-1/PFKFB3, and PKM2 and their expression characteristics in this autoimmune disease, and systematically assess the association between the glycolytic rate-limiting enzymes and RA from a molecular level. Moreover, we highlight HK-II, PFK-1/PFKFB3, and PKM2 as potential targets for the clinical treatment of RA. There is great potential to develop new anti-rheumatic therapies through safe inhibition or overexpression of glycolysis rate-limiting enzymes.
Collapse
Affiliation(s)
- Jianlin Zuo
- Department of Orthopeadics, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jinshuo Tang
- Department of Orthopeadics, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Meng Lu
- Department of Nursing, The First Bethune Hospital of Jilin University, Changchun, China
| | - Zhongsheng Zhou
- Department of Orthopeadics, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yang Li
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Hao Tian
- Department of Orthopeadics, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Enbo Liu
- Department of Orthopeadics, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Baoying Gao
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Te Liu
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Pu Shao
- Department of Orthopeadics, China-Japan Union Hospital of Jilin University, Changchun, China
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
12
|
Montes-Casado M, Ojeda G, Criado G, Rojo JM, Portolés P. The PI-3-Kinase P110α Catalytic Subunit of T Lymphocytes Modulates Collagen-Induced Arthritis. Int J Mol Sci 2021; 22:6405. [PMID: 34203838 PMCID: PMC8232790 DOI: 10.3390/ijms22126405] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 06/11/2021] [Indexed: 12/21/2022] Open
Abstract
The phosphatidylinositol 3-kinase (PI3K) family of enzymes plays a determinant role in inflammation and autoimmune responses. However, the implication of the different isoforms of catalytic subunits in these processes is not clear. Rheumatoid arthritis (RA) is a chronic, systemic autoimmune inflammatory disease that entails innate and adaptive immune response elements in which PI3K is a potential hub for immune modulation. In a mouse transgenic model with T-cell-specific deletion of p110α catalytic chain (p110α-/-ΔT), we show the modulation of collagen-induced arthritis (CIA) by this isoform of PI3K. In established arthritis, p110α-/-ΔT mice show decreased prevalence of illness than their control siblings, higher IgG1 titers and lower levels of IL-6 in serum, together with decreased ex vivo Collagen II (CII)-induced proliferation, IL-17A secretion and proportion of naive T cells in the lymph nodes. In a pre-arthritis phase, at 13 days post-Ag, T-cell-specific deletion of p110α chain induced an increased, less pathogenic IgG1/IgG2a antibodies ratio; changes in the fraction of naive and effector CD4+ subpopulations; and an increased number of CXCR5+ T cells in the draining lymph nodes of the p110α-/-ΔT mice. Strikingly, T-cell blasts in vitro obtained from non-immunized p110α-/-ΔT mice showed an increased expression of CXCR5, CD44 and ICOS surface markers and defective ICOS-induced signaling towards Akt phosphorylation. These results, plus the accumulation of cells in the lymph nodes in the early phase of the process, could explain the diminished illness incidence and prevalence in the p110α-/-ΔT mice and suggests a modulation of CIA by the p110α catalytic chain of PI3K, opening new avenues of intervention in T-cell-directed therapies to autoimmune diseases.
Collapse
Affiliation(s)
- María Montes-Casado
- Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, 28220 Madrid, Spain; (M.M.-C.); (G.O.)
| | - Gloria Ojeda
- Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, 28220 Madrid, Spain; (M.M.-C.); (G.O.)
| | - Gabriel Criado
- Grupo de Enfermedades Inflamatorias y Autoinmunes, Instituto de Investigación Hospital 12 de Octubre (i+12), 28041 Madrid, Spain;
| | - José M. Rojo
- Centro de Investigaciones Biológicas Margarita Salas, Departamento de Biomedicina Molecular, Consejo Superior de Investigaciones Científicas (CSIC), 28040 Madrid, Spain
| | - Pilar Portolés
- Centro Nacional de Microbiología, Instituto de Salud Carlos III (ISCIII), Majadahonda, 28220 Madrid, Spain; (M.M.-C.); (G.O.)
- Presidencia, Consejo Superior de Investigaciones Científicas (CSIC), 28006 Madrid, Spain
| |
Collapse
|
13
|
Wang T, Jiao Y, Zhang X. Immunometabolic Pathways and Its Therapeutic Implication in Autoimmune Diseases. Clin Rev Allergy Immunol 2020; 60:55-67. [PMID: 33179144 DOI: 10.1007/s12016-020-08821-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/02/2020] [Indexed: 02/08/2023]
Abstract
Autoimmune diseases (AIDs) are characterized with aberrant immune responses and their respective signaling pathways controlling cell differentiation, death, and survival. Cell metabolism is also an indispensable biochemical process that provides the very fundamental energy and materials. Accumulating evidences implicate that metabolism pathways have critical roles in determining the function of different immune subsets. Mechanisms of how immunometabolism participate in the pathogenesis of AIDs were also under intensive exploration. Here, in this review, we summarize the metabolic features of immune cells in AIDs and also the individual function of immunometabolism pathways, including glucose metabolism and tricarboxylic acid (TCA) cycle, in the setting of AIDs, mainly focusing on the potential targets for intervention. We also review studies that explore the intervention strategies targeting key molecules of metabolic pathways, such as mammalian target of rapamycin (mTOR), AMP-activated protein kinase (AMPK), and hypoxia-inducible factor 1a (HIF1a), in systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA). The highlight of this review is to provide a comprehensive summary of the status quo of immunometabolism studies in AIDs and the potential translatable drug targets.
Collapse
Affiliation(s)
- Tingting Wang
- Department of Medical Research Center, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100730, China.,Clinical Immunology Centre, Medical Epigenetics Research Centre, State Key Laboratory of Difficult and Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing , 100730, China.,State Key Laboratory of Difficult, Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing , 100730, China
| | - Yuhao Jiao
- Clinical Immunology Centre, Medical Epigenetics Research Centre, State Key Laboratory of Difficult and Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing , 100730, China.,Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College; The Ministry of Education Key Laboratory, Beijing , 100730, China
| | - Xuan Zhang
- Clinical Immunology Centre, Medical Epigenetics Research Centre, State Key Laboratory of Difficult and Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing , 100730, China. .,Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College; The Ministry of Education Key Laboratory, Beijing , 100730, China. .,State Key Laboratory of Difficult, Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing , 100730, China.
| |
Collapse
|
14
|
Masoumi M, Mehrabzadeh M, Mahmoudzehi S, Mousavi MJ, Jamalzehi S, Sahebkar A, Karami J. Role of glucose metabolism in aggressive phenotype of fibroblast-like synoviocytes: Latest evidence and therapeutic approaches in rheumatoid arthritis. Int Immunopharmacol 2020; 89:107064. [PMID: 33039953 DOI: 10.1016/j.intimp.2020.107064] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/10/2020] [Accepted: 09/30/2020] [Indexed: 12/12/2022]
Abstract
Glucose metabolism is considerably increased in inflamed joints of rheumatoid arthritis (RA) patients at early stages. Fibroblast-like synoviocytes (FLSs) activation and subsequent joint damage are linked with metabolic alterations, especially glucose metabolism. It has been shown that glucose metabolism is elevated in aggressive phenotype of FLS cells. In this regard, glycolytic blockers are able to reduce aggressiveness of the FLS cells resulting in decreased joint damage in various arthritis models. Besides, metabolic changes in immune and non-immune cells such as FLS can provide important targets for therapeutic intervention. Glycolytic enzymes such as hexokinase 2 (HK2), phosphofructo-2-kinase/fructose-2,6-bisphosphatase (PFKFB), and phosphoglycerate kinase (PGK) play essential roles in aggressive behavior of FLS cells. It has been documented that the HK2 enzyme is significantly upregulated in RA FLS cells, compared with osteoarthritis (OA) FLS cells. The HK2 is expressed in a few tissues and upregulated in the inflamed synovium of RA patients that makes it a potential target for RA treatment. Furthermore, HK2 has different roles in each cellular compartment, which offers another level of specificity and provides a specific target to reduce deleterious effects of inhibiting the enzyme in RA without affecting glycolysis in normal cells. Thus, targeting the HK2 enzyme might be an attractive potential selective target for arthritis therapy and safer than global glycolysis inhibition. Therefore, this review was aimed to summarize the current knowledge about glucose metabolism of FLS cells and suggest novel biomarkers, which are potential candidates for RA treatment.
Collapse
Affiliation(s)
- Maryam Masoumi
- Clinical Research Development Center, Shahid Beheshti Hospital, Qom University of Medical Sciences, Qom, Iran
| | - Mohsen Mehrabzadeh
- Department of Medical Biochemistry, Tehran University of Medical Sciences, Tehran, Iran
| | - Salman Mahmoudzehi
- Department of Medical Laboratory Sciences, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Mohammad Javad Mousavi
- Department of Hematology, Faculty of Allied Medicine, Bushehr University of Medical Sciences, Bushehr, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sirous Jamalzehi
- Department of Medical Laboratory Sciences, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland; Halal Research Center of IRI, FDA, Tehran, Iran.
| | - Jafar Karami
- Department of Laboratory Sciences, Khomein University of Medical Sciences, Khomein, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
15
|
Takahashi D, Hoshina N, Kabumoto Y, Maeda Y, Suzuki A, Tanabe H, Isobe J, Yamada T, Muroi K, Yanagisawa Y, Nakamura A, Fujimura Y, Saeki A, Ueda M, Matsumoto R, Asaoka H, Clarke JM, Harada Y, Umemoto E, Komatsu N, Okada T, Takayanagi H, Takeda K, Tomura M, Hase K. Microbiota-derived butyrate limits the autoimmune response by promoting the differentiation of follicular regulatory T cells. EBioMedicine 2020; 58:102913. [PMID: 32711255 PMCID: PMC7387783 DOI: 10.1016/j.ebiom.2020.102913] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 07/07/2020] [Accepted: 07/09/2020] [Indexed: 12/18/2022] Open
Abstract
Background Rheumatoid arthritis (RA) is a chronic debilitating autoimmune disorder with a high prevalence, especially in industrialized countries. Dysbiosis of the intestinal microbiota has been observed in RA patients. For instance, new-onset untreated RA (NORA) is associated with the underrepresentation of the Clostridium cluster XIVa, including Lachnospiraceae, which are major butyrate producers, although the pathological relevance has remained obscure. Follicular regulatory T (TFR) cells play critical regulatory roles in the pathogenesis of autoimmune diseases, including RA. Reduced number of circulating TFR cells has been associated with the elevation of autoantibodies and disease severity in RA. However, the contribution of commensal microbe-derived butyrate in controlling TFR cell differentiation remains unknown. Methods We examined the contribution of microbe-derived butyrate in controlling autoimmune arthritis using collagen-induced arthritis (CIA) and SKG arthritis models. We phenotyped autoimmune responses in the gut-associated lymphoid tissues (GALT) in the colon and joint-draining lymph nodes in the CIA model. We developed an in vitro CXCR5+Bcl-6+Foxp3+ TFR (iTFR) cell culture system and examined whether butyrate promotes the differentiation of iTFR cells. Findings Microbe-derived butyrate suppressed the development of autoimmune arthritis. The immunization of type II collagen (CII) caused hypertrophy of the GALT in the colon by amplifying the GC reaction prior to the onset of the CIA. Butyrate mitigated these pathological events by promoting TFR cell differentiation. Butyrate directly induced the differentiation of functional TFR cells in vitro by enhancing histone acetylation in TFR cell marker genes. This effect was attributed to histone deacetylase (HDAC) inhibition by butyrate, leading to histone hyperacetylation in the promoter region of the TFR-cell marker genes. The adoptive transfer of the butyrate-treated iTFR cells reduced CII-specific autoantibody production and thus ameliorated the symptoms of arthritis. Interpretation Accordingly, microbiota-derived butyrate serves as an environmental cue to enhance TFR cells, which suppress autoantibody production in the systemic lymphoid tissue, eventually ameliorating RA. Our findings provide mechanistic insights into the link between the gut environment and RA risk. Funding This work was supported by 10.13039/100009619AMED-Crest (16gm1010004h0101, 17gm1010004h0102, 18gm1010004h0103, and 19gm1010004s0104 to KH), the Japan Society for the Promotion of Science (JP17KT0055, JP16H01369, and JP18H04680 to KH; JP17K15734 to DT), Keio University Special Grant-in-Aid for Innovative Collaborative Research Projects (KH), Keio Gijuku Fukuzawa Memorial Fund for the Advancement of Education and Research (DT), the SECOM Science and Technology Foundation (KH), the Cell Science Research Foundation (KH), the Mochida Memorial Foundation for Medical and Pharmaceutical Research (DT), the Suzuken Memorial Foundation (KH and DT), the Takeda Science Foundation (KH and DT), The Science Research Promotion Fund, and The Promotion and Mutual Aid Corporation for Private Schools of Japan (KH).
Collapse
Affiliation(s)
- Daisuke Takahashi
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Sciences, Keio University, Minato-ku, Tokyo105-8512, Japan
| | - Naomi Hoshina
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Sciences, Keio University, Minato-ku, Tokyo105-8512, Japan
| | - Yuma Kabumoto
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Sciences, Keio University, Minato-ku, Tokyo105-8512, Japan
| | - Yuichi Maeda
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka565-0871, Japan
| | - Akari Suzuki
- Laboratory for Autoimmune Diseases, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Kanagawa230-0045, Japan
| | - Hiyori Tanabe
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Sciences, Keio University, Minato-ku, Tokyo105-8512, Japan
| | - Junya Isobe
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Sciences, Keio University, Minato-ku, Tokyo105-8512, Japan
| | - Takahiro Yamada
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Sciences, Keio University, Minato-ku, Tokyo105-8512, Japan
| | - Kisara Muroi
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Sciences, Keio University, Minato-ku, Tokyo105-8512, Japan
| | - Yuto Yanagisawa
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Sciences, Keio University, Minato-ku, Tokyo105-8512, Japan
| | - Atsuo Nakamura
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Sciences, Keio University, Minato-ku, Tokyo105-8512, Japan; Dairy Science and Technology Institute, Kyodo Milk Industry Co. Ltd., Nishitama, Tokyo190-0182, Japan
| | - Yumiko Fujimura
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Sciences, Keio University, Minato-ku, Tokyo105-8512, Japan
| | - Aiko Saeki
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Sciences, Keio University, Minato-ku, Tokyo105-8512, Japan
| | - Mizuki Ueda
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi, Osaka584-8540, Japan
| | - Ryohtaroh Matsumoto
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Sciences, Keio University, Minato-ku, Tokyo105-8512, Japan
| | - Hanako Asaoka
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Sciences, Keio University, Minato-ku, Tokyo105-8512, Japan
| | - Julie M Clarke
- Preventative Health National Research Flagship, CSIRO Food and Nutritional Sciences, Adelaide, South Australia5000, Australia
| | - Yohsuke Harada
- Laboratory of Pharmaceutical Immunology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba278-8510, Japan
| | - Eiji Umemoto
- Department of Microbiology and Immunology, Graduate School of Medicine, WPI Immunology Frontier Research Center (IFReC), Osaka University, Suita, Osaka565-0871, Japan
| | - Noriko Komatsu
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo113-0033, Japan
| | - Takaharu Okada
- Laboratory for Tissue Dynamics, RIKEN IMS, Yokohama, Kanagawa230-0045, Japan
| | - Hiroshi Takayanagi
- Department of Microbiology and Immunology, Graduate School of Medicine, WPI Immunology Frontier Research Center (IFReC), Osaka University, Suita, Osaka565-0871, Japan
| | - Kiyoshi Takeda
- Laboratory of Pharmaceutical Immunology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba278-8510, Japan
| | - Michio Tomura
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi, Osaka584-8540, Japan
| | - Koji Hase
- Division of Biochemistry, Faculty of Pharmacy and Graduate School of Pharmaceutical Sciences, Keio University, Minato-ku, Tokyo105-8512, Japan; International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo (IMSUT), Minato-ku, Tokyo108-8639, Japan.
| |
Collapse
|
16
|
Abstract
Rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE) are relatively common autoimmune diseases, often considered prototypic examples for how protective immunity switches to destructive immunity. The autoantigens recognized in RA and SLE are distinct, clinical manifestations are partially overlapping. A shared feature is the propensity of the adaptive immune system to respond inappropriately, with T cell hyper-responsiveness a pinnacle pathogenic defect. Upon antigen recognition, T cells mobilize a multi-pranged metabolic program, enabling them to massively expand and turn into highly mobile effector cells. Current evidence supports that T cells from patients with RA or SLE adopt metabolic programs different from healthy T cells, in line with the concept that autoimmune effector functions rely on specified pathways of energy sensing, energy generation and energy utilization. Due to misrouting of the energy sensor AMPK, RA T cells have a defect in balancing catabolic and anabolic processes and deviate towards a cell-building program. They supply biosynthetic precursors by shunting glucose away from glycolytic breakdown towards the pentose phosphate pathway and upregulate lipogenesis, enabling cellular motility and tissue invasiveness. Conversely, T cells from SLE patients are committed to high glycolytic flux, overusing the mitochondrial machinery and imposing oxidative stress. Typically, disease-relevant effector functions in SLE are associated with inappropriate activation of the key metabolic regulator mTORC1. Taken together, disease-specific metabolic signatures in RA and SLE represent vulnerabilities that are therapeutically targetable to suppress pathogenic immune responses.
Collapse
Affiliation(s)
- Bowen Wu
- School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Jörg J. Goronzy
- School of Medicine, Stanford University, Stanford, CA 94305, USA
- Department of Medicine, Palo Alto Veterans Administration Healthcare System, Palo Alto, CA 94304, USA
| | - Cornelia M. Weyand
- School of Medicine, Stanford University, Stanford, CA 94305, USA
- Department of Medicine, Palo Alto Veterans Administration Healthcare System, Palo Alto, CA 94304, USA
| |
Collapse
|
17
|
Zezina E, Sercan‐Alp O, Herrmann M, Biesemann N. Glucose transporter 1 in rheumatoid arthritis and autoimmunity. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2020; 12:e1483. [DOI: 10.1002/wsbm.1483] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 01/28/2020] [Accepted: 01/29/2020] [Indexed: 12/21/2022]
Affiliation(s)
- Ekaterina Zezina
- Sanofi R&D Immunology and Inflammation Therapeutic Area Type 1/17 Inflammation and Arthritis Cluster, Industriepark Hoechst Frankfurt am Main Germany
| | - Oezen Sercan‐Alp
- Sanofi R&D Immunology and Inflammation Therapeutic Area Type 1/17 Inflammation and Arthritis Cluster, Industriepark Hoechst Frankfurt am Main Germany
| | - Matthias Herrmann
- Sanofi R&D Immunology and Inflammation Therapeutic Area Type 1/17 Inflammation and Arthritis Cluster, Industriepark Hoechst Frankfurt am Main Germany
| | - Nadine Biesemann
- Sanofi R&D Immunology and Inflammation Therapeutic Area Type 1/17 Inflammation and Arthritis Cluster, Industriepark Hoechst Frankfurt am Main Germany
| |
Collapse
|
18
|
Panneton V, Chang J, Witalis M, Li J, Suh W. Inducible T‐cell co‐stimulator: Signaling mechanisms in T follicular helper cells and beyond. Immunol Rev 2019; 291:91-103. [DOI: 10.1111/imr.12771] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 04/30/2019] [Indexed: 12/30/2022]
Affiliation(s)
- Vincent Panneton
- IRCM (Institut de recherches cliniques de Montréal) Montreal Quebec Canada
- Department of Microbiology, Infectiology, and Immunology University of Montreal Montreal Quebec Canada
| | - Jinsam Chang
- IRCM (Institut de recherches cliniques de Montréal) Montreal Quebec Canada
- Molecular Biology Program University of Montreal Montreal Quebec Canada
| | - Mariko Witalis
- IRCM (Institut de recherches cliniques de Montréal) Montreal Quebec Canada
- Molecular Biology Program University of Montreal Montreal Quebec Canada
| | - Joanna Li
- IRCM (Institut de recherches cliniques de Montréal) Montreal Quebec Canada
- Department of Microbiology and Immunology McGill University Montreal Quebec Canada
| | - Woong‐Kyung Suh
- IRCM (Institut de recherches cliniques de Montréal) Montreal Quebec Canada
- Department of Microbiology, Infectiology, and Immunology University of Montreal Montreal Quebec Canada
- Molecular Biology Program University of Montreal Montreal Quebec Canada
- Department of Microbiology and Immunology McGill University Montreal Quebec Canada
| |
Collapse
|
19
|
Okano T, Saegusa J, Takahashi S, Ueda Y, Morinobu A. Immunometabolism in rheumatoid arthritis. Immunol Med 2018; 41:89-97. [PMID: 30938274 DOI: 10.1080/25785826.2018.1531186] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Recent studies have revealed a relationship between cellular metabolism and cell function in immune cells. Cellular metabolism not only provides supplemental ATP, but also supports dynamic changes in cell proliferation and differentiation. For example, T cells exhibit subset-specific metabolic profiles, and require certain types of metabolism for their functions. Determining the metabolic profiles that support inflammatory immune responses may lead to novel treatment strategies for chronic inflammatory diseases such as rheumatoid arthritis (RA). However, the mechanisms by which metabolism modulates cell function have been unclear. Recent studies have begun to unveil unexpected non-metabolic functions for metabolic enzymes in the context of inflammation, including roles in signaling and gene regulation. Here we describe recent findings related to immunometabolism, the metabolome of RA patients, and the metabolically independent functions of glycolytic enzymes. We discuss how metabolic processes impact immune cells, especially T cells and fibroblast like synoviocytes, which are considered the orchestrators of autoimmune arthritis.
Collapse
Affiliation(s)
- Takaichi Okano
- a Clinical Laboratory , Kobe University Hospital , Kobe , Japan.,b Department of Rheumatology and Clinical Immunology , Kobe University Graduate School of Medicine , Kobe , Japan
| | - Jun Saegusa
- a Clinical Laboratory , Kobe University Hospital , Kobe , Japan.,b Department of Rheumatology and Clinical Immunology , Kobe University Graduate School of Medicine , Kobe , Japan
| | - Soshi Takahashi
- c Center for Rheumatic Diseases , Shinko Hospital , Kobe , Japan
| | - Yo Ueda
- b Department of Rheumatology and Clinical Immunology , Kobe University Graduate School of Medicine , Kobe , Japan
| | - Akio Morinobu
- b Department of Rheumatology and Clinical Immunology , Kobe University Graduate School of Medicine , Kobe , Japan
| |
Collapse
|
20
|
Zhang M, Wu Y, Bastian D, Iamsawat S, Chang J, Daenthanasanmak A, Nguyen HD, Schutt S, Dai M, Chen F, Suh WK, Yu XZ. Inducible T-Cell Co-Stimulator Impacts Chronic Graft-Versus-Host Disease by Regulating Both Pathogenic and Regulatory T Cells. Front Immunol 2018; 9:1461. [PMID: 29988391 PMCID: PMC6023972 DOI: 10.3389/fimmu.2018.01461] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 06/12/2018] [Indexed: 01/22/2023] Open
Abstract
The incidence of chronic graft-versus-host disease (cGVHD) is on the rise and still the major cause of morbidity and mortality among patients after allogeneic hematopoietic stem cell transplantation (HCT). Both donor T and B cells contribute to the pathogenesis of cGVHD. Inducible T-cell co-stimulator (ICOS), a potent co-stimulatory receptor, plays a key role in T-cell activation and differentiation. Yet, how ICOS regulates the development of cGVHD is not well understood. Here, we investigated the role of ICOS in cGVHD pathogenesis using mice with germline or regulatory T cell (Treg)-specific ICOS deficiency. The recipients of ICOS−/− donor grafts had reduced cGVHD compared with wild-type controls. In recipients of ICOS−/− donor grafts, we observed significant reductions in donor T follicular helper (Tfh), Th17, germinal center B-cell, and plasma cell differentiation, coupled with lower antibody production. Interestingly, Tregs, including follicular regulatory T (Tfr) cells, were also impaired in the absence of ICOS. Using ICOS conditional knockout specific for Foxp3+ cells, we found that ICOS was indispensable for optimal survival and homeostasis of induced Tregs during cGVHD. Furthermore, administration of anti-ICOS alleviated cGVHD severity via suppressing T effector cells without affecting Treg generation. Taken together, ICOS promotes T- and B-cell activation and differentiation, which can promote cGVHD development; however, ICOS is critical for the survival and homeostasis of iTregs, which can suppress cGVHD. Hence, ICOS balances the development of cGVHD and could offer a potential target after allo-HCT in the clinic.
Collapse
Affiliation(s)
- Mengmeng Zhang
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, China
| | - Yongxia Wu
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States
| | - David Bastian
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States
| | - Supinya Iamsawat
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States
| | - Jinsam Chang
- Institut de Recherches Cliniques de Montréal (IRCM), Montreal, QC, Canada
| | - Anusara Daenthanasanmak
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States
| | - Hung D Nguyen
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States
| | - Steven Schutt
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States
| | - Min Dai
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fangping Chen
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, China
| | - Woong-Kyung Suh
- Institut de Recherches Cliniques de Montréal (IRCM), Montreal, QC, Canada
| | - Xue-Zhong Yu
- Department of Microbiology and Immunology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, United States.,Department of Medicine, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|