1
|
Christodoulou M, Moysidou E, Lioulios G, Stai S, Lazarou C, Xochelli A, Fylaktou A, Stangou M. T-Follicular Helper Cells and Their Role in Autoimmune Diseases. Life (Basel) 2025; 15:666. [PMID: 40283219 PMCID: PMC12028949 DOI: 10.3390/life15040666] [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: 03/11/2025] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 04/29/2025] Open
Abstract
T-follicular helper (Tfh) cells, a specialized subset of CD4+ cells, are the immune mediators connecting cellular and humoral immunity, as they lead B-cell proliferation within germinal centers, and orchestrate their response, including activation, class switching, and production of a diverse array of high-affinity antibodies. Their interactions with B cells is regulated by a wide complex of transcriptional and cytokine-driven pathways. A major contribution of Tfh cells to autoimmune diseases is through their production of cytokines, particularly IL-21, which supports the proliferation and differentiation of autoreactive B cells. Elevated levels of circulating Tfh-like cells and IL-21 have been observed in patients with systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA) correlating strongly with disease severity and autoantibody levels. The feedback loop between Tfh cells and IL-21 or other signal pathways, such as Bcl-6, ICOS, and PD-1, not only sustains Tfh cell function but also drives the continuous expansion of autoreactive B cells, leading to chronic inflammation through the production of high-affinity pathogenic autoantibodies. By understanding these interactions, Tfh pathways may serve as potential therapeutic targets, with IL-21, ICOS, and PD1 blockades emerging as promising innovative therapeutic strategies to manage autoimmune diseases. Although a variety of studies have been conducted investigating the role of Tfh cells in SLE and RA, this review aims to reveal the gap in the literature regarding the role of such subpopulations in the pathogenesis of other autoimmune diseases, such as Anca-associated vasculitis (AAV), and express the need to conduct similar studies. Tfh cell-related biomarkers can be used to assess disease activity and transform autoimmune disease treatment, leading to more personalized and effective care for patients with chronic autoimmune conditions.
Collapse
Affiliation(s)
- Michalis Christodoulou
- School of Medicine, Aristotle University of Thessaloniki, Department of Nephrology, Hippokration Hospital, Konstantinoupoleos 49, 54642 Thessaloniki, Greece; (M.C.); (E.M.); (G.L.); (S.S.); (C.L.)
| | - Eleni Moysidou
- School of Medicine, Aristotle University of Thessaloniki, Department of Nephrology, Hippokration Hospital, Konstantinoupoleos 49, 54642 Thessaloniki, Greece; (M.C.); (E.M.); (G.L.); (S.S.); (C.L.)
| | - Georgios Lioulios
- School of Medicine, Aristotle University of Thessaloniki, Department of Nephrology, Hippokration Hospital, Konstantinoupoleos 49, 54642 Thessaloniki, Greece; (M.C.); (E.M.); (G.L.); (S.S.); (C.L.)
| | - Stamatia Stai
- School of Medicine, Aristotle University of Thessaloniki, Department of Nephrology, Hippokration Hospital, Konstantinoupoleos 49, 54642 Thessaloniki, Greece; (M.C.); (E.M.); (G.L.); (S.S.); (C.L.)
| | - Christina Lazarou
- School of Medicine, Aristotle University of Thessaloniki, Department of Nephrology, Hippokration Hospital, Konstantinoupoleos 49, 54642 Thessaloniki, Greece; (M.C.); (E.M.); (G.L.); (S.S.); (C.L.)
| | - Aliki Xochelli
- Department of Immunology, National Histocompatibility Center, Hippokration General Hospital, 54642 Thessaloniki, Greece; (A.X.); (A.F.)
| | - Asimina Fylaktou
- Department of Immunology, National Histocompatibility Center, Hippokration General Hospital, 54642 Thessaloniki, Greece; (A.X.); (A.F.)
| | - Maria Stangou
- School of Medicine, Aristotle University of Thessaloniki, Department of Nephrology, Hippokration Hospital, Konstantinoupoleos 49, 54642 Thessaloniki, Greece; (M.C.); (E.M.); (G.L.); (S.S.); (C.L.)
| |
Collapse
|
2
|
Satoh-Kanda Y, Nakayamada S, Kubo S, Yamagata K, Nawata A, Tanaka H, Kosaka S, Kanda R, Yu S, Fujita Y, Sonomoto K, Tanaka Y. Modifying T cell phenotypes using TYK2 inhibitor and its implications for the treatment of systemic lupus erythematosus. RMD Open 2024; 10:e003991. [PMID: 38871479 PMCID: PMC11177773 DOI: 10.1136/rmdopen-2023-003991] [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: 12/11/2023] [Accepted: 05/31/2024] [Indexed: 06/15/2024] Open
Abstract
OBJECTIVES The tuning effects of JAK/TYK2 inhibitors on the imbalance between T follicular helper (Tfh) and T regulatory (Treg) cells, related to systemic lupus erythematosus (SLE) pathogenesis, were investigated using human peripheral blood samples. METHODS Peripheral blood mononuclear cells from untreated patients with SLE and healthy controls were analysed. Tfh1 cells were identified in nephritis tissue, and the effect of Tfh1 cells on B-cell differentiation was examined by coculturing naïve B cells with Tfh1 cells. RESULTS Tfh1 cell numbers were increased in the peripheral blood of patients, and activated Treg cell counts were decreased relative to Tfh1 cell counts. This imbalance in the Tfh to Treg ratio was remarkably pronounced in cases of lupus nephritis, especially in types III and IV active nephritis. Immunohistochemistry revealed Tfh1 cell infiltration in lupus nephritis tissues. Co-culture of Tfh1 cells (isolated from healthy individuals) with naïve B cells elicited greater induction of T-bet+ B cells than controls. In JAK/TYK2-dependent STAT phosphorylation assays using memory CD4+ T cells, IL-12-induced STAT1/4 phosphorylation and Tfh1 cell differentiation were inhibited by both JAK and TYK2 inhibitors. However, phosphorylation of STAT5 by IL-2 and induction of Treg cell differentiation by IL-2+TGFβ were inhibited by JAK inhibitors but not by TYK2 inhibitors, suggesting that TYK2 does not mediate the IL-2 signalling pathway. CONCLUSIONS Tfh1 cells can induce T-bet+ B cell production and may contribute to SLE pathogenesis-associated processes. TYK2 inhibitor may fine-tune the immune imbalance by suppressing Tfh1 differentiation and maintaining Treg cell differentiation, thereby preserving IL-2 signalling, unlike other JAK inhibitors.
Collapse
Affiliation(s)
- Yurie Satoh-Kanda
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Fukuoka, Japan
| | - Shingo Nakayamada
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Fukuoka, Japan
| | - Satoshi Kubo
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Fukuoka, Japan
- Department of Molecular Targeted Therapies (DMTT), University of Occupational and Environmental Health, Japan, Kitakyushu, Fukuoka, Japan
| | - Kaoru Yamagata
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Fukuoka, Japan
| | - Aya Nawata
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Fukuoka, Japan
- Department of Pathology and Oncology, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Hiroaki Tanaka
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Fukuoka, Japan
| | - Shunpei Kosaka
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Fukuoka, Japan
- Department of Pathology and Oncology, University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Ryuichiro Kanda
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Fukuoka, Japan
| | - Shan Yu
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Fukuoka, Japan
- Department of Pediatrics, Shenyang Women's and Children's Hospital, Shenyang, Liaoning, China
| | - Yuya Fujita
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Fukuoka, Japan
| | - Koshiro Sonomoto
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Fukuoka, Japan
| | - Yoshiya Tanaka
- The First Department of Internal Medicine, University of Occupational and Environmental Health, Japan, Kitakyushu, Fukuoka, Japan
| |
Collapse
|
3
|
Kan AKC, Tang WT, Li PH. Helper T cell subsets: Development, function and clinical role in hypersensitivity reactions in the modern perspective. Heliyon 2024; 10:e30553. [PMID: 38726130 PMCID: PMC11079302 DOI: 10.1016/j.heliyon.2024.e30553] [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: 01/14/2024] [Revised: 04/28/2024] [Accepted: 04/29/2024] [Indexed: 05/12/2024] Open
Abstract
Helper T cells are traditionally classified into T helper 1 (TH1) and T helper 2 (TH2). The more recent discoveries of T helper 17 (TH17), follicular helper T cells (TFH) and regulatory T cells (Treg) enhanced our understanding on the mechanisms of immune function and hypersensitivity reactions, which shaped the modern perspective on the function and role of these different subsets of helper T cells in hypersensitivity reactions. Each subset of helper T cells has characteristic roles in different types of hypersensitivity reactions, hence giving rise to the respective characteristic clinical manifestations. The roles of helper T cells in allergic contact dermatitis (TH1-mediated), drug rash with eosinophilia and systemic symptoms (DRESS) syndrome (TH2-mediated), and acute generalised exanthematous pustulosis (AGEP) (TH17-mediated) are summarised in this article, demonstrating the correlation between the type of helper T cell involved and the clinical features. TFH plays crucial roles in antibody class-switch recombination; they may be implicated in antibody-mediated hypersensitivity reactions, but further research is warranted to delineate their exact pathogenic roles. The helper T cell subsets and their specific cytokine profiles implicated in different hypersensitivity reactions could be potential treatment targets by biologics, but more clinical trials are warranted to establish their clinical effectiveness.
Collapse
Affiliation(s)
- Andy Ka Chun Kan
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, Hong Kong Special Administrative Region of China
| | - Wang Tik Tang
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, Hong Kong Special Administrative Region of China
| | - Philip H. Li
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, Hong Kong Special Administrative Region of China
| |
Collapse
|
4
|
Harrison J, Newland SA, Jiang W, Giakomidi D, Zhao X, Clement M, Masters L, Corovic A, Zhang X, Drago F, Ma M, Ozsvar Kozma M, Yasin F, Saady Y, Kothari H, Zhao TX, Shi GP, McNamara CA, Binder CJ, Sage AP, Tarkin JM, Mallat Z, Nus M. Marginal zone B cells produce 'natural' atheroprotective IgM antibodies in a T cell-dependent manner. Cardiovasc Res 2024; 120:318-328. [PMID: 38381113 PMCID: PMC10939463 DOI: 10.1093/cvr/cvae027] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/10/2023] [Accepted: 12/12/2023] [Indexed: 02/22/2024] Open
Abstract
AIMS The adaptive immune response plays an important role in atherosclerosis. In response to a high-fat/high-cholesterol (HF/HC) diet, marginal zone B (MZB) cells activate an atheroprotective programme by regulating the differentiation and accumulation of 'poorly differentiated' T follicular helper (Tfh) cells. On the other hand, Tfh cells activate the germinal centre response, which promotes atherosclerosis through the production of class-switched high-affinity antibodies. We therefore investigated the direct role of Tfh cells and the role of IL18 in Tfh differentiation in atherosclerosis. METHODS AND RESULTS We generated atherosclerotic mouse models with selective genetic deletion of Tfh cells, MZB cells, or IL18 signalling in Tfh cells. Surprisingly, mice lacking Tfh cells had increased atherosclerosis. Lack of Tfh not only reduced class-switched IgG antibodies against oxidation-specific epitopes (OSEs) but also reduced atheroprotective natural IgM-type anti-phosphorylcholine (PC) antibodies, despite no alteration of natural B1 cells. Moreover, the absence of Tfh cells was associated with an accumulation of MZB cells with substantially reduced ability to secrete antibodies. In the same manner, MZB cell deficiency in Ldlr-/- mice was associated with a significant decrease in atheroprotective IgM antibodies, including natural anti-PC IgM antibodies. In humans, we found a positive correlation between circulating MZB-like cells and anti-OSE IgM antibodies. Finally, we identified an important role for IL18 signalling in HF/HC diet-induced Tfh. CONCLUSION Our findings reveal a previously unsuspected role of MZB cells in regulating atheroprotective 'natural' IgM antibody production in a Tfh-dependent manner, which could have important pathophysiological and therapeutic implications.
Collapse
Affiliation(s)
- James Harrison
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Stephen A Newland
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Wei Jiang
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Despoina Giakomidi
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Xiaohui Zhao
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Marc Clement
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- Laboratory for Vascular Translational Sciences (LVTS), Université de Paris, INSERM U1148, Paris, France
| | - Leanne Masters
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Andrej Corovic
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Xian Zhang
- Department of Medicine, Brigham and Woman’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Fabrizio Drago
- Division of Cardiovascular Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Marcella Ma
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, UK
| | - Maria Ozsvar Kozma
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Froher Yasin
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Yuta Saady
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Hema Kothari
- Division of Cardiovascular Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Tian X Zhao
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Guo-Ping Shi
- Department of Medicine, Brigham and Woman’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Coleen A McNamara
- Division of Cardiovascular Medicine, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Andrew P Sage
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Jason M Tarkin
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Ziad Mallat
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- PARCC Inserm U970, Universite de Paris, Paris, France
| | - Meritxell Nus
- Department of Medicine, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| |
Collapse
|
5
|
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
|
6
|
Wu C, Jiang ML, Pang T, Zhang CJ. T Cell Subsets and Immune Homeostasis. Methods Mol Biol 2024; 2782:39-63. [PMID: 38622391 DOI: 10.1007/978-1-0716-3754-8_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2024]
Abstract
T cells are a heterogeneous group of cells that can be classified into different subtypes according to different classification methods. The body's immune system has a highly complex and effective regulatory network that allows for the relative stability of immune system function. Maintaining proper T cell homeostasis is essential for promoting protective immunity and limiting autoimmunity and tumor formation. Among the T cell family members, more and more T cell subsets have gradually been characterized. In this chapter, we summarize the functions of some key T cell subsets and their impact on immune homeostasis.
Collapse
Affiliation(s)
- Chuyu Wu
- Department of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing, China
| | - Mei-Ling Jiang
- Department of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Tao Pang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, China Pharmaceutical University, Nanjing, China
| | - Cun-Jin Zhang
- Department of Neurology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| |
Collapse
|
7
|
Nakayamada S, Tanaka Y. Immune Phenotype as a Biomarker for Systemic Lupus Erythematosus. Biomolecules 2023; 13:960. [PMID: 37371540 DOI: 10.3390/biom13060960] [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: 03/31/2023] [Revised: 05/29/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
The treatment of rheumatoid arthritis was revolutionized with the use of molecular-targeted drugs that target immunoregulatory molecules. The success of treatment with these drugs prompted the development of molecular-targeted drugs for systemic lupus erythematosus. However, systemic lupus erythematosus is a disease with high heterogeneous immune abnormalities, and diverse cells or molecules can be treatment targets. Thus, the identification of subpopulations based on immune abnormalities is essential for the development of effective treatment. One analytical method used to identify subpopulations is the immunophenotyping of peripheral blood samples of patients. This analysis evaluates the validity of target molecules for peripheral blood immune cell subsets, which are expected to be developed as biomarkers for precision medicine in which appropriate treatment targets are set for each subpopulation.
Collapse
Affiliation(s)
- Shingo Nakayamada
- The First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Japan, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu 807-8555, Fukuoka, Japan
| | - Yoshiya Tanaka
- The First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Japan, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu 807-8555, Fukuoka, Japan
| |
Collapse
|
8
|
Chen JY, Tian XY, Wei SS, Xu W, Pan RR, Chen LL, Chen LD, Nan LH, Wang QQ, Ma XQ, Huang MQ. Magnolol as STAT3 inhibitor for treating multiple sclerosis by restricting Th17 cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 117:154917. [PMID: 37301184 DOI: 10.1016/j.phymed.2023.154917] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 05/21/2023] [Accepted: 06/02/2023] [Indexed: 06/12/2023]
Abstract
OBJECTIVE Multiple sclerosis (MS) is an immune disease in the central nervous system (CNS) associated with Th17 cells. Moreover, STAT3 initiates Th17 cell differentiation and IL-17A expression through facilitating RORγt in MS. Here, we reported that magnolol, isolated from Magnolia officinalis Rehd. Et Wils, was regarded as a candidate for MS treatment verified by both in vitro and in vivo studies. METHODS In vivo, experimental autoimmune encephalomyelitis (EAE) model in mice was employed to evaluate the alleviation of magnolol on myeloencephalitis. In vitro, FACS assay was employed to evaluate the effect of magnolol on Th17 and Treg cell differentiation and IL-17A expression; network pharmacology-based study was applied to probe the involved mechanisms; western blotting, immunocytochemistry, and luciferase reporter assay was used to further confirm the regulation of magnolol on JAK/STATs signaling pathway; surface plasmon resonance (SPR) assay and molecular docking were applied to manifest affinity with STAT3 and binding sites; overexpression of STAT3 was employed to verify whether magnolol attenuates IL-17A through STAT3 signaling pathway. RESULTS In vivo, magnolol alleviated loss of body weight and severity of EAE mice; magnolol improved lesions in spinal cords and attenuated CD45 infiltration, and serum cytokines levels; correspondingly, magnolol focused on inhibiting Th17 differentiation and IL-17A expression in splenocyte of EAE mice; moreover, magnolol selectively inhibited p-STAT3(Y705) and p-STAT4(Y693) of both CD4+ and CD8+ T cells in splenocyte of EAE mice. In vitro, magnolol selectively inhibited Th17 differentiation and IL-17A expression without impact on Treg cells; network pharmacology-based study revealed that magnolol perhaps diminished Th17 cell differentiation through regulating STAT family members; western blotting further confirmed that magnolol inhibited p-JAK2(Y1007) and selectively antagonized p-STAT3(Y705) and slightly decreased p-STAT4(Y693); magnolol antagonized both STAT3 nucleus location and transcription activity; magnolol had a high affinity with STAT3 and the specific binding site perhaps to be at SH2 domain; overexpression of STAT3 resulted in failed inhibition of magnolol on IL-17A. CONCLUSION Magnolol selectively inhibited Th17 differentiation and cytokine expression through selectively blocking of STAT3 resulting in decreased the ratio of Th17/Treg cells for treating MS, suggesting that the potential of magnolol for treating MS as novel STAT3 inhibitor.
Collapse
Affiliation(s)
- Jian-Yu Chen
- Department of Pharmacology, School of Pharmacy, Fujian University of Traditional Chinese Medicine, No.1, Hua Tuo Road, Min Hou Shang Jie, Fuzhou 350122, China
| | - Xiao-Yun Tian
- Department of Pharmacology, School of Pharmacy, Fujian University of Traditional Chinese Medicine, No.1, Hua Tuo Road, Min Hou Shang Jie, Fuzhou 350122, China
| | - Shan-Shan Wei
- School of Pharmacy, Second Military Medical University, No.325, Guo He Road, Shanghai 30025, China
| | - Wen Xu
- Department of Pharmacology, School of Pharmacy, Fujian University of Traditional Chinese Medicine, No.1, Hua Tuo Road, Min Hou Shang Jie, Fuzhou 350122, China
| | - Rong-Rong Pan
- Seventh People's Hospital of Shanghai University of Traditional Chinese Medicine, No. 358, Datong road, Pudong New Area, Shanghai 200137, China
| | - Lin-Lin Chen
- School of Pharmacy, Second Military Medical University, No.325, Guo He Road, Shanghai 30025, China
| | - Lang-Dong Chen
- School of Pharmacy, Second Military Medical University, No.325, Guo He Road, Shanghai 30025, China
| | - Li-Hong Nan
- Department of Pharmacology, School of Pharmacy, Fujian University of Traditional Chinese Medicine, No.1, Hua Tuo Road, Min Hou Shang Jie, Fuzhou 350122, China
| | - Qian-Qian Wang
- Medical College, Dalian University, Dalian 116622, China.
| | - Xue-Qin Ma
- Department of Pharmaceutical Analysis, School of Pharmacy, Key Laboratory of Hui Ethnic Medicine Modernization, Ministry of Education, Ningxia Medical University, 1160 Shenli Street, Yinchuan 750004, China.
| | - Ming-Qing Huang
- Department of Pharmacology, School of Pharmacy, Fujian University of Traditional Chinese Medicine, No.1, Hua Tuo Road, Min Hou Shang Jie, Fuzhou 350122, China.
| |
Collapse
|
9
|
Nakayamada S, Tanaka Y. Development of targeted therapies in IgG4-related disease. Mod Rheumatol 2023; 33:266-270. [PMID: 35983919 DOI: 10.1093/mr/roac096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 08/07/2022] [Accepted: 08/16/2022] [Indexed: 11/14/2022]
Abstract
IgG4-related disease (IgG4-RD) is a systemic disease characterized by high serum IgG4 levels, infiltration of lymphocytes and IgG4-positive plasma cells into affected tissues, and subsequent fibrosis, forming mass, nodular, and thickened lesions in organs. Although glucocorticoids (GCs) are the first-line treatment for IgG4-RD, the disease often relapses during dose reduction or after discontinuation of GC. Long-term treatment with GC is associated with adverse effects such as infection, osteoporosis, and atherosclerosis. Therefore, there is an urgent need to develop a treatment strategy that specifically addresses the pathogenesis of IgG4-RD. As immunocompetent cells and immune-related molecules involved in the pathogenesis of IgG4-RD are increasingly being identified, there is a growing demand for new molecular-targeted drugs that target them. In particular, favourable results have been reported for drugs that target B cells, such as anti-cluster of differentiation (CD)20 and anti-CD19 antibodies. In addition, clinical trials are underway for new therapeutic agents, such as anti-signalling lymphocytic activation molecule family 7 antibodies that target T cells and other cells.
Collapse
Affiliation(s)
- Shingo Nakayamada
- The First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Fukuoka, Japan
| | - Yoshiya Tanaka
- The First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Fukuoka, Japan
| |
Collapse
|
10
|
Jia X, Zhu H, Jiang Q, Gu J, Yu S, Chi X, Wang R, Shan Y, Jiang H, Ma X. Identification of key genes and imbalance of immune cell infiltration in immunoglobulin A associated vasculitis nephritis by integrated bioinformatic analysis. Front Immunol 2023; 14:1087293. [PMID: 37026011 PMCID: PMC10070996 DOI: 10.3389/fimmu.2023.1087293] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 03/02/2023] [Indexed: 04/08/2023] Open
Abstract
Background IgAV, the most common systemic vasculitis in childhood, is an immunoglobulin A-associated immune complex-mediated disease and its underlying molecular mechanisms are not fully understood. This study attempted to identify differentially expressed genes (DEGs) and find dysregulated immune cell types in IgAV to find the underlying pathogenesis for IgAVN. Methods GSE102114 datasets were obtained from the Gene Expression Omnibus (GEO) database to identify DEGs. Then, the protein-protein interaction (PPI) network of the DEGs was constructed using the STRING database. And key hub genes were identified by cytoHubba plug-in, performed functional enrichment analyses and followed by verification using PCR based on patient samples. Finally, the abundance of 24 immune cells were detected by Immune Cell Abundance Identifier (ImmuCellAI) to estimate the proportions and dysregulation of immune cell types within IgAVN. Result A total of 4200 DEGs were screened in IgAVN patients compared to Health Donor, including 2004 upregulated and 2196 downregulated genes. Of the top 10 hub genes from PPI network, STAT1, TLR4, PTEN, UBB, HSPA8, ATP5B, UBA52, and CDC42 were verified significantly upregulated in more patients. Enrichment analyses indicated that hub genes were primarily enriched in Toll-like receptor (TLR) signaling pathway, nucleotide oligomerization domain (NOD)-like receptor signaling pathway, and Th17 signaling pathways. Moreover, we found a diversity of immune cells in IgAVN, consisting mainly of T cells. Finally, this study suggests that the overdifferentiation of Th2 cells, Th17 cells and Tfh cells may be involved in the occurrence and development of IgAVN. Conclusion We screened out the key genes, pathways and maladjusted immune cells and associated with the pathogenesis of IgAVN. The unique characteristics of IgAV-infiltrating immune cell subsets were confirmed, providing new insights for future molecular targeted therapy and a direction for immunological research on IgAVN.
Collapse
Affiliation(s)
- Xianxian Jia
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, China
| | - Hua Zhu
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, China
| | - Qinglian Jiang
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, China
- Department of General Pediatrics, Zhongshan City People’s Hospital, Guangzhou, China
| | - Jia Gu
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, China
| | - Shihan Yu
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, China
| | - Xuyang Chi
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, China
| | - Rui Wang
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, China
| | - Yu Shan
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Hong Jiang
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, China
| | - Xiaoxue Ma
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, China
- Department of Microbiology & Immunology and Pediatrics, Dalhousie University, Halifax, NS, Canada
- *Correspondence: Xiaoxue Ma,
| |
Collapse
|
11
|
Wakui M, Uwamino Y, Yatabe Y, Nakagawa T, Sakai A, Kurafuji T, Shibata A, Tomita Y, Noguchi M, Tanabe A, Arai T, Ohno A, Yokota H, Uno S, Yamasawa W, Sato Y, Ikeda M, Yoshimura A, Hasegawa N, Saya H, Murata M. Assessing anti-SARS-CoV-2 cellular immunity in 571 vaccines by using an IFN-γ release assay. Eur J Immunol 2022; 52:1961-1971. [PMID: 36250411 PMCID: PMC9874394 DOI: 10.1002/eji.202249794] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 09/23/2022] [Accepted: 10/12/2022] [Indexed: 01/27/2023]
Abstract
Memory T cell responses have been analyzed only in small cohorts of COVID-19 vaccines. Herein, we aimed to assess anti-SARS-CoV-2 cellular immunity in a large cohort using QuantiFERON assays, which are IFN-γ release assays (IGRAs) based on short-term whole blood culture. The study included 571 individuals receiving the viral spike (S) protein-expressing BNT162b2 mRNA vaccine. QuantiFERON assays revealed antigen-specific IFN-γ production in most individuals 8 weeks after the second dose. Simultaneous flow cytometric assays to detect T cells expressing activation-induced markers (AIMs) performed for 28 randomly selected individuals provided data correlating with the QuantiFERON data. Simultaneous IFN-γ enzyme-linked immunospot and AIM assays for another subset of 31 individuals, based on short-term peripheral blood mononuclear cell culture, also indicated a correlation between IFN-γ production and AIM positivity. These observations indicated the acquisition of T cell memory responses and supported the usability of IGRAs to assess cellular immunity. The QuantiFERON results were weakly correlated with serum IgG titers against the receptor-binding domain of the S protein and were associated with pre-vaccination infection and adverse reactions after the second dose. The present study revealed cellular immunity after COVID-19 vaccination, providing insights into the effects and adverse reactions of vaccination.
Collapse
Affiliation(s)
- Masatoshi Wakui
- Department of Laboratory MedicineKeio University School of MedicineShinjuku‐kuTokyoJapan
| | - Yoshifumi Uwamino
- Department of Laboratory MedicineKeio University School of MedicineShinjuku‐kuTokyoJapan,Department of Infectious DiseasesKeio University School of MedicineShinjuku‐kuTokyoJapan
| | - Yoko Yatabe
- Clinical LaboratoryKeio University HospitalShinjuku‐kuTokyoJapan
| | | | - Akiko Sakai
- Clinical LaboratoryKeio University HospitalShinjuku‐kuTokyoJapan
| | | | - Ayako Shibata
- Department of Laboratory MedicineKeio University School of MedicineShinjuku‐kuTokyoJapan
| | - Yukari Tomita
- Department of Laboratory MedicineKeio University School of MedicineShinjuku‐kuTokyoJapan
| | - Masayo Noguchi
- Clinical LaboratoryKeio University HospitalShinjuku‐kuTokyoJapan
| | - Akiko Tanabe
- Clinical LaboratoryKeio University HospitalShinjuku‐kuTokyoJapan
| | - Tomoko Arai
- Clinical LaboratoryKeio University HospitalShinjuku‐kuTokyoJapan
| | - Akemi Ohno
- Clinical LaboratoryKeio University HospitalShinjuku‐kuTokyoJapan
| | - Hiromitsu Yokota
- Clinical LaboratoryKeio University HospitalShinjuku‐kuTokyoJapan
| | - Shunsuke Uno
- Department of Infectious DiseasesKeio University School of MedicineShinjuku‐kuTokyoJapan
| | - Wakako Yamasawa
- Department of Laboratory MedicineKeio University School of MedicineShinjuku‐kuTokyoJapan
| | - Yasunori Sato
- Department of Epidemiology and Preventive MedicineKeio University School of MedicineShinjuku‐kuTokyoJapan
| | - Mari Ikeda
- Department of Microbiology and ImmunologyKeio University School of MedicineShinjuku‐kuTokyoJapan
| | - Akihiko Yoshimura
- Department of Microbiology and ImmunologyKeio University School of MedicineShinjuku‐kuTokyoJapan
| | - Naoki Hasegawa
- Department of Infectious DiseasesKeio University School of MedicineShinjuku‐kuTokyoJapan
| | - Hideyuki Saya
- Division of Gene RegulationInstitute for Advanced Medical ResearchKeio University School of MedicineShinjuku‐kuTokyoJapan
| | - Mitsuru Murata
- Department of Laboratory MedicineKeio University School of MedicineShinjuku‐kuTokyoJapan
| |
Collapse
|
12
|
Chi X, Gu J, Ma X. Characteristics and Roles of T Follicular Helper Cells in SARS-CoV-2 Vaccine Response. Vaccines (Basel) 2022; 10:vaccines10101623. [PMID: 36298488 PMCID: PMC9611968 DOI: 10.3390/vaccines10101623] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 09/26/2022] [Accepted: 09/26/2022] [Indexed: 11/17/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccination is critical to controlling the coronavirus disease 2019 (COVID-19) pandemic. However, a weak response to the vaccine and insufficient persistence of specific antibodies may threaten the global impact of mass vaccination campaigns. This study summarizes the internal factors of the body that affect the effectiveness of the SARS-CoV-2 vaccine. T follicular helper (Tfh) cells support germinal center B cells to produce vaccine-specific immunoglobulins. A reduction in the Tfh cell number and a shift in the subset phenotypes caused by multiple factors may impair the production and persistence of high-affinity antibodies. Besides efficacy differences caused by the different types of vaccines, the factors that affect vaccine effectiveness by intervening in the Tfh cell response also include age-related defects, the polarity of the body microenvironment, repeated immunization, immunodeficiency, and immunosuppressive treatments. Assessing the phenotypic distribution and activation levels of Tfh cell subsets after vaccination is helpful in predicting vaccine responses and may identify potential targets for improving vaccine effectiveness.
Collapse
Affiliation(s)
- Xuyang Chi
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang 110001, China
| | - Jia Gu
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang 110001, China
| | - Xiaoxue Ma
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang 110001, China
- Department of Microbiology & Immunology and Pediatrics, Dalhousie University, and Canadian Center for Vaccinology, IWK Health Centre, Halifax, NS B3K 6R8, Canada
- Correspondence: ; Tel.: +86-024-83282527
| |
Collapse
|
13
|
Jin X, Chen J, Wu J, Lu Y, Li B, Fu W, Wang W, Cui D. Aberrant expansion of follicular helper T cell subsets in patients with systemic lupus erythematosus. Front Immunol 2022; 13:928359. [PMID: 36119056 PMCID: PMC9478104 DOI: 10.3389/fimmu.2022.928359] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 08/15/2022] [Indexed: 11/25/2022] Open
Abstract
Objective Systemic lupus erythematosus (SLE) is a chronic and complex autoimmune disease characterized by multiple autoantibodies, resulting in multiple organ and tissue damages. These pathogenic autoantibodies produced by B cells are closely correlated with follicular helper T (Tfh) cell subsets that play a fundamental role in the pathogenesis of SLE. The aim of the present study was to study the phenotype and role of circulating Tfh (cTfh) cell subsets and associated B cell subpopulations in active and inactive SLE patients. Methods Thirty SLE outpatients and 24 healthy controls (HCs) were enrolled in this study. The frequency of cTfh cell and B cell subsets in peripheral blood mononuclear cells (PBMCs) and the plasma levels of eight cytokines were determined by flow cytometry, and plasma IL-21 levels were measured by ELISA. Meanwhile, we used MRL/lpr mice as the model of SLE to research the alterations of Tfh cells in the thymus and spleen of mice. Results Frequencies of CD4+CXCR5+CD45RA-effector cTfh cells, PD1+cTfh, PD1+ICOS+cTfh, PD1+cTfh1, PD1+cTfh2, PD1+cTfh17, and PD1+ICOS+cTfh1 cells as well as plasmablasts showed significant differences among HC, active and inactive SLE patients. Moreover, cytokines typically associated with cTfh cells, including IL-6 and IL-21, were elevated in active SLE patients compared to inactive SLE patients and HCs. Additionally, a positive correlation was observed between PD1+ICOS+ cTfh or PD1+ICOS+ cTfh1 cell frequencies and plasmablasts or IL-21 levels, as well as between plasmablasts. We also found PD1+ICOS+ Tfh cells expansion in both thymus and spleen of MRL/lpr mice, accompanied by increased frequencies in B cells and plasmablasts, meanwhile, cTfh1which expressing IFN-γ was increased in the peripheral blood of MRL/lpr mice. Conclusion Tfh cell subsets and plasmablasts may play a fundamental role in the pathogenesis of SLE and may provide potential targets for therapeutic interventions for SLE.
Collapse
Affiliation(s)
- Xin Jin
- Department of Clinical Laboratory, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Jia Chen
- Department of Nephrology Research, The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jian Wu
- Department of Clinical Laboratory, Suzhou Hospital Affiliated to Nanjing Medical University, Suzhou, China
| | - Ying Lu
- Department of Rheumatology, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Baohua Li
- Department of Clinical Laboratory, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Wenning Fu
- Department of Rheumatology, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Wei Wang
- Department of Clinical Laboratory, Tongde Hospital of Zhejiang Province, Hangzhou, China
- *Correspondence: Dawei Cui, ; Wei Wang,
| | - Dawei Cui
- Department of Blood Transfusion, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Dawei Cui, ; Wei Wang,
| |
Collapse
|
14
|
Govender M, Hopkins FR, Göransson R, Svanberg C, Shankar EM, Hjorth M, Nilsdotter-Augustinsson Å, Sjöwall J, Nyström S, Larsson M. T cell perturbations persist for at least 6 months following hospitalization for COVID-19. Front Immunol 2022; 13:931039. [PMID: 36003367 PMCID: PMC9393525 DOI: 10.3389/fimmu.2022.931039] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 07/14/2022] [Indexed: 01/08/2023] Open
Abstract
COVID-19 is being extensively studied, and much remains unknown regarding the long-term consequences of the disease on immune cells. The different arms of the immune system are interlinked, with humoral responses and the production of high-affinity antibodies being largely dependent on T cell immunity. Here, we longitudinally explored the effect COVID-19 has on T cell populations and the virus-specific T cells, as well as neutralizing antibody responses, for 6-7 months following hospitalization. The CD8+ TEMRA and exhausted CD57+ CD8+ T cells were markedly affected with elevated levels that lasted long into convalescence. Further, markers associated with T cell activation were upregulated at inclusion, and in the case of CD69+ CD4+ T cells this lasted all through the study duration. The levels of T cells expressing negative immune checkpoint molecules were increased in COVID-19 patients and sustained for a prolonged duration following recovery. Within 2-3 weeks after symptom onset, all COVID-19 patients developed anti-nucleocapsid IgG and spike-neutralizing IgG as well as SARS-CoV-2-specific T cell responses. In addition, we found alterations in follicular T helper (TFH) cell populations, such as enhanced TFH-TH2 following recovery from COVID-19. Our study revealed significant and long-term alterations in T cell populations and key events associated with COVID-19 pathogenesis.
Collapse
Affiliation(s)
- Melissa Govender
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Francis R. Hopkins
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Robin Göransson
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- Department of Clinical Immunology and Transfusion Medicine, and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Cecilia Svanberg
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Esaki M. Shankar
- Infection Biology, Department of Life Sciences, Central University of Tamil Nadu, Thiruvarur, India
| | - Maria Hjorth
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- Department of Clinical Immunology and Transfusion Medicine, and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Åsa Nilsdotter-Augustinsson
- Divison of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Johanna Sjöwall
- Divison of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Sofia Nyström
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- Department of Clinical Immunology and Transfusion Medicine, and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Marie Larsson
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- *Correspondence: Marie Larsson,
| |
Collapse
|
15
|
Wu Y, Wu C, Che Y, Zhang T, Dai C, Nguyễn AD, Duan K, Huang Y, Li N, Zhou H, Wan X, Wang Y, Lei H, Hao P, Li C, Wu Y. Effects of Glycyrrhiza Polysaccharides on Chickens' Intestinal Health and Homeostasis. Front Vet Sci 2022; 9:891429. [PMID: 35647094 PMCID: PMC9134109 DOI: 10.3389/fvets.2022.891429] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 04/05/2022] [Indexed: 01/20/2023] Open
Abstract
The overuse of antibiotics in poultry farming causes the accumulation of drug residue in animals' bodies and the occurrence of antibiotic-resistant bacteria, which not only compromise animals' health but ultimately endanger human health. Thus, there is an urgent need for a novel poultry feed additive to substitute for excessive antibiotics. Glycyrrhiza polysaccharides (GPS) derived from Chinese licorice have shown promising immunomodulatory effects in previous studies. The present study investigated the pharmacological effects of GPS on poultry intestines to assess whether it can be used as a feed additive. The results show that GPS can increase production of sIgA, promote the secretion activity of goblet cells, alter the gut microbial composition and lead to changes in short-chain fatty acids. GPS also elevated both Th1 and Th2 immune responses by facilitating the expression of IL-2, IL-4, IL-1β, and IFN-γ while increasing the proportion of both CD4+ and CD8+ cells in the intestine. Moreover, the results of 16S rRNA gene sequencing showed that GPS could significantly change intestinal microbiota composition in the intestine, evidenced by the increased proportion of Bacteroides, Butyricicoccus and Eisenbergiella, as well as a decreased portion of Erysipelatoclostridium, leading to a healthier intestinal microbiota composition for the host. Taken together, it can be concluded that GPS is safe to use as a novel feed additive that can be used as an alternative to prophylactic antibiotics in poultry feeding.
Collapse
Affiliation(s)
- Yu Wu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Chenyang Wu
- Beijing Key Laboratory of Traditional Chinese Veterinary Medicine, Beijing University of Agriculture, Beijing, China
| | - Yanyun Che
- Engineering Laboratory for National Healthcare Theories and Products of Yunnan Province, College of Pharmaceutical Science, Yunnan University of Chinese Medicine, Kunming, China
| | - Tao Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Chen Dai
- College of Life Sciences, Experimental Teaching Center of Life Science, Nanjing Agricultural University, Nanjing, China
| | - Audrey D. Nguyễn
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, CA, United States
| | - Kun Duan
- China Tobacco Henan Industrial Co., Ltd., Zhengzhou, China
| | - Yanyu Huang
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, CA, United States
| | - Nannan Li
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Hui Zhou
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xin Wan
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yuedi Wang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Hongjun Lei
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Ping Hao
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Caiyue Li
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yi Wu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- *Correspondence: Yi Wu ;
| |
Collapse
|
16
|
Liu Y, Hu X, Hu X, Yu L, Ji H, Li W, Cai Y, Cheng G, Jiang Y. T follicular helper cells improve the response of patients with chronic hepatitis B to interferon by promoting HBsAb production. J Gastroenterol 2022; 57:30-45. [PMID: 34988689 DOI: 10.1007/s00535-021-01840-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 11/26/2021] [Indexed: 02/04/2023]
Abstract
BACKGROUND AND AIMS Hepatitis B surface antigen (HBsAg) seroconversion is considered the optimal outcome of the treatment of chronic hepatitis B virus (HBV) infection. In this study, we aimed to determine the cellular and molecular mechanisms by which pegylated interferon alpha (PEG-IFN-α) improves the seroconversion rate in patients with chronic hepatitis B (CHB). METHODS Flow cytometry was performed using circulating T follicular helper (TFH) cells from 15 healthy individuals and 45 patients with CHB presenting different treatment responses [complete response group (CRG), incomplete response group (ICRG), and nonresponse group (NRG)] to the standard 48-week regimen of PEG-IFN-α monotherapy to examine the significance of circulating TFH cells in the therapeutic response of patients with CHB to PEG-IFN-α. In addition, the capacities of different TFH subsets to activate B cells and stimulate IgG production were assessed by performing coculture experiments. RESULTS Longitudinal analysis revealed specific and significant increases in the numbers of CD40L+CD4+CXCR5+ TFH cells in the CRG compared with the NRG and ICRG. According to the results of in vitro coculture experiments, blocking CD40-CD40L signaling, but not ICOS-ICOSL signaling, specifically inhibits B-cell activation and IgG production. HBV may impair TFH cell function by enhancing inhibitory regulatory T-cell activity. Transcriptome analysis further revealed the upregulation of CD40L, but not of ICOS, in TFH cells isolated from the CRG. CONCLUSIONS TFH cells, particularly those with CD40L expression, stimulate B-cell differentiation and improve the HBsAg seroconversion rate in patients with CHB treated with PEG-IFN-α monotherapy.
Collapse
Affiliation(s)
- Yong Liu
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Xintong Hu
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Xiaoli Hu
- Department of Infectious Disease, Heilongjiang Provincial Hospital, Harbin, China
| | - Lei Yu
- Department of Infectious Disease, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Huifan Ji
- Department of Hepatology, The First Hospital of Jilin University, Changchun, China
| | - Wanyu Li
- Department of Hepatology, The First Hospital of Jilin University, Changchun, China
| | - Yanjun Cai
- Department of Hepatology, The First Hospital of Jilin University, Changchun, China
| | - Genhong Cheng
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, 130021, China.,Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA, 90095, USA
| | - Yanfang Jiang
- Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, 130021, China. .,Key Laboratory of Zoonosis Research, Ministry of Education, The First Hospital of Jilin University, Changchun, 130021, China.
| |
Collapse
|
17
|
OUP accepted manuscript. Rheumatology (Oxford) 2022; 61:4547-4557. [DOI: 10.1093/rheumatology/keac112] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 02/11/2022] [Indexed: 11/14/2022] Open
|
18
|
Cui D, Tang Y, Jiang Q, Jiang D, Zhang Y, Lv Y, Xu D, Wu J, Xie J, Wen C, Lu L. Follicular Helper T Cells in the Immunopathogenesis of SARS-CoV-2 Infection. Front Immunol 2021; 12:731100. [PMID: 34603308 PMCID: PMC8481693 DOI: 10.3389/fimmu.2021.731100] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 09/01/2021] [Indexed: 12/21/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is a serious infectious disease that has led to a global pandemic with high morbidity and mortality. High-affinity neutralizing antibody is important for controlling infection, which is closely regulated by follicular helper T (Tfh) cells. Tfh cells play a central role in promoting germinal center reactions and driving cognate B cell differentiation for antibody secretion. Available studies indicate a close relationship between virus-specific Tfh cell-mediated immunity and SARS-CoV-2 infection progression. Although several lines of evidence have suggested that Tfh cells contribute to the control of SARS-CoV-2 infection by eliciting neutralizing antibody productions, further studies are needed to elucidate Tfh-mediated effector mechanisms in anti-SARS-CoV-2 immunity. Here, we summarize the functional features and roles of virus-specific Tfh cells in the immunopathogenesis of SARS-CoV-2 infection and in COVID-19 vaccines, and highlight the potential of targeting Tfh cells as therapeutic strategy against SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Dawei Cui
- Department of Blood Transfusion, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuan Tang
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong, Hong Kong, Hong Kong, SAR China.,Chongqing International Institute for Immunology, Chongqing, China
| | - Qi Jiang
- Department of Blood Transfusion, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China
| | - Daixi Jiang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yun Zhang
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yan Lv
- Department of Blood Transfusion, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dandan Xu
- Department of Blood Transfusion, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jian Wu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jue Xie
- Department of Blood Transfusion, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chengping Wen
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Liwei Lu
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong, Hong Kong, Hong Kong, SAR China.,Chongqing International Institute for Immunology, Chongqing, China
| |
Collapse
|
19
|
Nakayamada S, Tanaka Y. Clinical relevance of T follicular helper cells in systemic lupus erythematosus. Expert Rev Clin Immunol 2021; 17:1143-1150. [PMID: 34469695 DOI: 10.1080/1744666x.2021.1976146] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION T helper cells regulate a variety of immune responses and are involved in the pathogenesis of infection, allergy and autoimmune diseases. T follicular helper (Tfh) cells, which induce B cell maturation, play an important role in the production of the extremely diverse autoantibodies found in systemic lupus erythematosus (SLE). AREA COVERED We provide an overview of the plasticity and diversity of Tfh cells in humans and their involvement in the pathology and pathogenesis of SLE. Our review outlines the potential of Tfh cells as a therapeutic target for SLE. EXPERT OPINION Tfh cells are involved in the pathogenesis of SLE based on their plasticity and diversity. Tfh cell differentiation and function are variably regulated by cytokines (IL-12, interferons, IL-2, etc), co-stimulatory molecules (ICOS, CD40L, OX40, etc), and intracellular signals (JAK-STAT, etc). Elucidation of the mechanisms underlying Tfh cell differentiation and function may lead to the development of new therapies for SLE.
Collapse
Affiliation(s)
- Shingo Nakayamada
- The First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yoshiya Tanaka
- The First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| |
Collapse
|