1
|
Nie J, Qin X, Tao X, Huang J. Exploring the molecular landscape of lymphocyte activation gene-3: A literature review. Medicine (Baltimore) 2024; 103:e39622. [PMID: 39331884 PMCID: PMC11441911 DOI: 10.1097/md.0000000000039622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 08/16/2024] [Indexed: 09/29/2024] Open
Abstract
Molecular structure and cellular distribution of lymphocyte activation gene-3 (LAG-3) have been studied extensively since 1990. However, several unresolved questions remain. It is well-established that LAG-3 plays a significant role in maintaining immune homeostasis. The presence of deficiencies in LAG-3 has been observed to be linked with autoimmune disorders, whereas the excessive expression of LAG-3 within the tumor microenvironment hinders immune responses, particularly those mediated by lymphocytes, thereby facilitating immune evasion. Consequently, investigations into these 2 aspects have become a prominent focus in both fundamental and clinical research. The objective of this review is to examine the functions and molecular characteristics of LAG-3, as well as its current clinical applications in the context of tumor immune escape and autoimmune disease. The ultimate aim is to explore and propose novel immune therapy approach.
Collapse
Affiliation(s)
- Jiaqi Nie
- Clinical Laboratory Center, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xue Qin
- Clinical Laboratory Center, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xiang Tao
- Clinical Laboratory Center, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jin Huang
- Clinical Laboratory Center, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
2
|
Wang F, Liu J. Regulating the lncRNA DSCR9/RPLP2/PI3K/AKT axis: an important mechanism of Xinfeng capsules in improving rheumatoid arthritis. Front Immunol 2024; 15:1465442. [PMID: 39376558 PMCID: PMC11456487 DOI: 10.3389/fimmu.2024.1465442] [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: 07/16/2024] [Accepted: 09/04/2024] [Indexed: 10/09/2024] Open
Abstract
Background Rheumatoid arthritis (RA) is a systemic autoimmune disease characterized by chronic and symmetrical polyarthritis. RA patients often experience inflammatory reaction and hypercoagulable state, which together affect the self-perception of patient (SPP). Currently, inhibiting inflammation and hypercoagulable state are common treatment methods for alleviating RA symptoms. Xinfeng Capsules (XFC) has a long history of treating RA, and can effectively improve the inflammatory response and hypercoagulable state of RA. However, the potential mechanisms have not yet been determined. Purpose and study design This study elucidated the action mechanism of XFC in RA inflammation and hypercoagulability through the lncDSCR9/RPLP2/PI3K/AKT axis. Results Clinical observations indicated that there was a strong link between XFC therapy and improvements in inflammatory and coagulation biomarkers, as well as SPP among RA patients. The subsequent network pharmacology analysis results identified the PI3K/AKT signaling pathway as a potential mediator for XFC treatment of RA. Furthermore, clinical validation and sequencing results revealed that lncRNA DSCR9 expression (a gene implicated in inflammation and coagulation) was negatively correlated with clinical markers of inflammation and coagulation, while positively correlated with SF-36 indicators. Notably, XFC treatment remarkably upregulated lncRNA DSCR9 expression and downregulated PI3K and AKT expressions, showing opposite expression trends to the untreated cases.The regulatory effect of XFC on the lncRNA DSCR9/RPLP2/PI3K/AKT axis in RA was investigated using techniques such as RNA pull-down assay, Western blot analysis, RT-PCR, and EdU assay. Moreover, the administration of the PI3K/AKT agonist RMH can counteract the effects of XFC on p-PI3K, p-AKT, inflammation, and hypercoagulability, reinforcing the role of pathway. Finally, animal studies utilizing HE staining and transmission electron microscopy (TEM) demonstrated that XFC notably decreased PI3K and AKT expressions in adjuvant-induced arthritis (AA) rats, mitigated inflammation and hypercoagulability, and enhanced the ultrastructure of synovial cells. These findings underscored the potential mechanisms of XFC in the treatment of RA. Conclusion Regulating the lncRNA DSCR9/RPLP2/PI3K/AKT axis may be an important mechanism by which XFC improved RA inflammatory response and hypercoagulable state.
Collapse
Affiliation(s)
- Fanfan Wang
- The First Affiliated Hospital of Anhui University of Chinese Medicine, First Clinical Medical College, Hefei, Anhui, China
- Department of Rheumatism Immunity, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Jian Liu
- Department of Rheumatism Immunity, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
| |
Collapse
|
3
|
Haham N, Zveik O, Rechtman A, Brill L, Vaknin-Dembinsky A. Altered immune co-inhibitory receptor expression and correlation of LAG-3 expression to disease severity in NMOSD. J Neuroimmunol 2024; 388:578289. [PMID: 38301597 DOI: 10.1016/j.jneuroim.2024.578289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/20/2023] [Accepted: 01/10/2024] [Indexed: 02/03/2024]
Abstract
Co-inhibitory receptors (CIR)s regulate T cell-mediated immune responses and growing evidence links co-inhibitory receptors to the progression of neuroimmunological diseases. We studied the expression levels of CIRs: TIM-3, TIGIT, PD-1 and LAG-3 in the peripheral blood mononuclear cells (PBMCs) of 30 patients with Neuromyelitis optica spectrum disorder (NMOSD), 11 Multiple sclerosis (MS) patients and 31 Healthy controls (HC). We found that the mRNA expression levels of TIM-3 were significantly increased in NMOSD compared with HC, and increased LAG-3 surface protein expression was also observed on T-cells of NMOSD patients. Moreover, we observed a negative correlation between LAG-3 expression and disease severity in NMOSD. Our findings suggest a protective effect of LAG-3 in the setting of NMOSD, and that the differential expression of CIRs observed in this study may play a role in the pathological process of NMOSD.
Collapse
Affiliation(s)
- Nitsan Haham
- Department of Neurology and Laboratory of Neuroimmunology and the Agnes-Ginges Center for Neurogenetics, Hadassah- Medical Center, Ein-Kerem, Faculty of Medicine, Hebrew University of Jerusalem, Ein-Karem, Jerusalem 91120, Israel.
| | - Omri Zveik
- Department of Neurology and Laboratory of Neuroimmunology and the Agnes-Ginges Center for Neurogenetics, Hadassah- Medical Center, Ein-Kerem, Faculty of Medicine, Hebrew University of Jerusalem, Ein-Karem, Jerusalem 91120, Israel
| | - Ariel Rechtman
- Department of Neurology and Laboratory of Neuroimmunology and the Agnes-Ginges Center for Neurogenetics, Hadassah- Medical Center, Ein-Kerem, Faculty of Medicine, Hebrew University of Jerusalem, Ein-Karem, Jerusalem 91120, Israel
| | - Livnat Brill
- Department of Neurology and Laboratory of Neuroimmunology and the Agnes-Ginges Center for Neurogenetics, Hadassah- Medical Center, Ein-Kerem, Faculty of Medicine, Hebrew University of Jerusalem, Ein-Karem, Jerusalem 91120, Israel
| | - Adi Vaknin-Dembinsky
- Department of Neurology and Laboratory of Neuroimmunology and the Agnes-Ginges Center for Neurogenetics, Hadassah- Medical Center, Ein-Kerem, Faculty of Medicine, Hebrew University of Jerusalem, Ein-Karem, Jerusalem 91120, Israel.
| |
Collapse
|
4
|
Qin L, Lin H, Zhu F, Wang J, Feng T, Xu T, Zhang G, Zhang X. CD4 +LAG3 +T cells are decreased in SSc-ILD and affect fibroblast mesenchymal transition by TGF-β3. iScience 2023; 26:108225. [PMID: 38025770 PMCID: PMC10661698 DOI: 10.1016/j.isci.2023.108225] [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: 05/08/2023] [Revised: 09/04/2023] [Accepted: 10/13/2023] [Indexed: 12/01/2023] Open
Abstract
Pulmonary fibrosis frequently occurs in rheumatic conditions, particularly systemic sclerosis-associated interstitial lung disease (SSc-ILD). The pathology involves cell transformation into interstitial structures and collagen accumulation. CD4+LAG3+T cells, known for immune inhibition, are relevant in autoimmunity. This study investigates CD4+LAG3+T cells in SSc-ILD. Clinical analysis revealed a correlation between CD4+LAG3+T cells and interleukin-6 (IL-6) and erythrocyte sedimentation rate (ESR). Using primary human lung fibroblasts (pHLFs) and murine bone marrow-derived macrophages (BMDMs), we showed that CD4+LAG3+T cells secreted TGF-β3 inhibits TGF-β1-induced mesenchymal transformation, modulates cellular function, and reduces collagen release. In mouse models, CD4+LAG3+T cells exhibited potential in alleviating bleomycin-induced pulmonary fibrosis. This study emphasizes CD4+LAG3+T cells' therapeutic promise against fibrosis and proposes their role as biomarkers.
Collapse
Affiliation(s)
- Linmang Qin
- Department of Rheumatology and Immunology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Haobo Lin
- Department of Rheumatology and Immunology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Fu Zhu
- Liuzhou Worker’s Hospital, Liuzhou 545007, China
| | - Jieying Wang
- Department of Rheumatology and Immunology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Tianxiao Feng
- Department of Rheumatology and Immunology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Ting Xu
- Department of Rheumatology and Immunology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Guangfeng Zhang
- Department of Rheumatology and Immunology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Xiao Zhang
- Department of Rheumatology and Immunology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| |
Collapse
|
5
|
Mueller AA, Sasaki T, Keegan JW, Nguyen JP, Griffith A, Horisberger AM, Licata T, Fieg E, Cao Y, Elahee M, Marks KE, Simmons DP, Briere LC, Cobban LA, Pallais JC, High FA, Walker MA, Linnoila JJ, Sparks JA, Holers VM, Costenbader KH, Sweetser DA, Krier JB, Loscalzo J, Lederer JA, Rao DA. High-dimensional immunophenotyping reveals immune cell aberrations in patients with undiagnosed inflammatory and autoimmune diseases. J Clin Invest 2023; 133:e169619. [PMID: 37874643 PMCID: PMC10721141 DOI: 10.1172/jci169619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2023] Open
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Daimon P. Simmons
- Department of Medicine
- Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | | | | | | | | | - Melissa A. Walker
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jenny J. Linnoila
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | - V. Michael Holers
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | | | | | | | | | | | | | | |
Collapse
|
6
|
Hu S, Tao Y, Hu F, Liu X. Diminished LAG3 + B cells correlate with exacerbated rheumatoid arthritis. Ann Med 2023; 55:2208373. [PMID: 37143367 PMCID: PMC10165927 DOI: 10.1080/07853890.2023.2208373] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/08/2023] [Accepted: 04/24/2023] [Indexed: 05/06/2023] Open
Abstract
BACKGROUND Lymphocyte activation gene-3 (LAG3) positive B cells have been identified as a novel regulatory B cell subset, while the role of LAG3+ B cells in the pathogenesis of rheumatoid arthritis (RA) remains elusive. MATERIALS AND METHODS Peripheral blood mononuclear cells (PBMCs) from RA, osteoarthritis (OA) patients and healthy volunteers were collected for flow cytometry staining of LAG3+ B cells. Their correlation with RA patient clinical and immunological features were analyzed. Moreover, the frequencies of LAG3+ B cells in collagen-induced arthritis (CIA) mice and naive mice were also detected. RESULTS A significant decrease of LAG3+ B cells was observed in RA patients as compared with healthy individuals and OA patients. Notably, the frequencies of LAG3+ B cells were negatively correlated with tender joint count (r = -0.4301, p = .0157) and DAS28-ESR (r = -0.4018, p = .025) in RA patients. In CIA mice, LAG3+ B cell frequencies were also decreased and negatively correlated with the CIA arthritis score. CONCLUSIONS Impairment of LAG3+ B cells potentially contributes to RA development. Reconstituting LAG3+ B cells might provide novel therapeutic strategies for the persistent disease.Key messagesLAG3+ B cells have been identified as a novel regulatory B cell subset. However, its role in the pathogenesis of RA remains unknown.This study revealed the decreased frequency of LAG3+ B cells in RA patients. Notably, LAG3+ B cells were negatively correlated with RA disease activity including the tender joint count and DAS28-ESR.In CIA mice, LAG3+ B cell frequencies were also decreased and negatively correlated with the CIA arthritis score.Reconstitution of LAG3+ B cells might provide novel therapeutic strategies for disease perpetuation.
Collapse
Affiliation(s)
- Suiyuan Hu
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Yuting Tao
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Fanlei Hu
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Xu Liu
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| |
Collapse
|
7
|
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
|
8
|
Aggarwal V, Workman CJ, Vignali DAA. LAG-3 as the third checkpoint inhibitor. Nat Immunol 2023; 24:1415-1422. [PMID: 37488429 PMCID: PMC11144386 DOI: 10.1038/s41590-023-01569-z] [Citation(s) in RCA: 103] [Impact Index Per Article: 51.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 06/19/2023] [Indexed: 07/26/2023]
Abstract
Lymphocyte activation gene 3 (LAG-3) is an inhibitory receptor that is highly expressed by exhausted T cells. LAG-3 is a promising immunotherapeutic target, with more than 20 LAG-3-targeting therapeutics in clinical trials and a fixed-dose combination of anti-LAG-3 and anti-PD-1 now approved to treat unresectable or metastatic melanoma. Although LAG-3 is widely recognized as a potent inhibitory receptor, important questions regarding its biology and mechanism of action remain. In this Perspective, we focus on gaps in the understanding of LAG-3 biology and discuss the five biggest topics of current debate and focus regarding LAG-3, including its ligands, signaling and mechanism of action, its cell-specific functions, its importance in different disease settings, and the development of novel therapeutics.
Collapse
Affiliation(s)
- Vaishali Aggarwal
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Creg J Workman
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Dario A A Vignali
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
| |
Collapse
|
9
|
Zhou X, Gu Y, Wang H, Zhou W, Zou L, Li S, Hua C, Gao S. From bench to bedside: targeting lymphocyte activation gene 3 as a therapeutic strategy for autoimmune diseases. Inflamm Res 2023:10.1007/s00011-023-01742-y. [PMID: 37314518 DOI: 10.1007/s00011-023-01742-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 01/12/2023] [Accepted: 05/12/2023] [Indexed: 06/15/2023] Open
Abstract
BACKGROUND Immune checkpoints negatively regulate immune response, thereby playing an important role in maintaining immune homeostasis. Substantial studies have confirmed that blockade or deficiency of immune checkpoint pathways contributes to the deterioration of autoimmune diseases. In this context, focusing on immune checkpoints might provide alternative strategies for the treatment of autoimmunity. Lymphocyte activation gene 3 (LAG3), as a member of immune checkpoint, is critical in regulating immune responses as manifested in multiple preclinical studies and clinical trials. Recent success of dual-blockade of LAG3 and programmed death-1 in melanoma also supports the notion that LAG3 is a crucial regulator in immune tolerance. METHODS We wrote this review article by searching the PubMed, Web of Science and Google Scholar databases. CONCLUSION In this review, we summarize the molecular structure and the action mechanisms of LAG3. Additionally, we highlight its roles in diverse autoimmune diseases and discuss how the manipulation of the LAG3 pathway can serve as a promising therapeutic strategy as well as its specific mechanism with the aim of filling the gaps from bench to bedside.
Collapse
Affiliation(s)
- Xueyin Zhou
- School of the 2nd Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yiming Gu
- School of the 2nd Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Huihong Wang
- School of the 2nd Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Wei Zhou
- School of the 2nd Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Lei Zou
- School of the 2nd Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Shuting Li
- School of the 2nd Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Chunyan Hua
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| | - Sheng Gao
- Laboratory Animal Center, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China.
| |
Collapse
|
10
|
Intranasal administration of abatacept enhances IL-35+ and IL-10+ producing Bregs in lung tissues of ovalbumin-sensitized asthmatic mice model. PLoS One 2022; 17:e0271689. [PMID: 36067164 PMCID: PMC9447931 DOI: 10.1371/journal.pone.0271689] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Accepted: 07/05/2022] [Indexed: 11/24/2022] Open
Abstract
Backgrounds Treating asthmatic rheumatoid arthritis patients with abatacept has been shown to associate with better control of asthma symptoms. However, the mechanism behind that is not investigated. Methods Ovalbumin (OVA)- sensitized BALB/c female mice were treated intranasally (IN) or intraperitoneally (IP) with abatacept 4 hrs before the OVA challenge. The effects of abatacept IN or IP on the lungs and blood levels of Tregs and Bregs and their production of immunosuppressive cytokines, were determined using FACS analysis and ELISA assay. Results Treating OVA- sensitized asthmatic mice model with abatacept, IN or IP, reduced lung inflammation. IN treatment with abatacept increased the frequency of IL-35 and IL-10 producing Bregs in the lung tissues to a higher level compared to IP treatment. Moreover, the frequency of lungs LAG3+ Tregs was significantly increased following treatment. This was also associated with a reduction in lung tissue and serum IL-17 levels of treated mice. Conclusions These results suggest that abatacept by enhancing IL-35+IL-10+ Bregs and LAG3+ Tregs might reverse IL-17 induced lung inflammation during asthma.
Collapse
|
11
|
Roodenrijs NMT, Welsing PMJ, van Roon J, Schoneveld JLM, van der Goes MC, Nagy G, Townsend MJ, van Laar JM. Mechanisms underlying DMARD inefficacy in difficult-to-treat rheumatoid arthritis: a narrative review with systematic literature search. Rheumatology (Oxford) 2022; 61:3552-3566. [PMID: 35238332 PMCID: PMC9434144 DOI: 10.1093/rheumatology/keac114] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/07/2022] [Accepted: 02/14/2022] [Indexed: 12/03/2022] Open
Abstract
Management of RA patients has significantly improved over the past decades. However, a substantial proportion of patients is difficult-to-treat (D2T), remaining symptomatic after failing biological and/or targeted synthetic DMARDs. Multiple factors can contribute to D2T RA, including treatment non-adherence, comorbidities and co-existing mimicking diseases (e.g. fibromyalgia). Additionally, currently available biological and/or targeted synthetic DMARDs may be truly ineffective ('true' refractory RA) and/or lead to unacceptable side effects. In this narrative review based on a systematic literature search, an overview of underlying (immune) mechanisms is presented. Potential scenarios are discussed including the influence of different levels of gene expression and clinical characteristics. Although the exact underlying mechanisms remain largely unknown, the heterogeneity between individual patients supports the assumption that D2T RA is a syndrome involving different pathogenic mechanisms.
Collapse
Affiliation(s)
- Nadia M T Roodenrijs
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht
| | - Paco M J Welsing
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht
| | - Joël van Roon
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht
| | | | - Marlies C van der Goes
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht
- Department of Rheumatology, Meander Medical Center, Amersfoort, The Netherlands
| | - György Nagy
- Department of Rheumatology & Clinical Immunology
- Department of Genetics, Cell and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Michael J Townsend
- Biomarker Discovery OMNI, Genentech Research & Early Development, South San Francisco, CA, USA
| | - Jacob M van Laar
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht
| |
Collapse
|
12
|
Hemmatzadeh M, Ahangar Parvin E, Mohammadi H, Azizi G, Shomali N, Jadidi-Niaragh F. The role of immune regulatory molecules in rheumatoid arthritis: Implication for etiopathogenesis and prospective for treatment. J Cell Physiol 2022; 237:3541-3553. [PMID: 35938550 DOI: 10.1002/jcp.30855] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 06/20/2022] [Accepted: 07/25/2022] [Indexed: 11/10/2022]
Abstract
Rheumatoid arthritis (RA) is considered an autoimmune chronic disorder and the most common inflammatory arthropathy. Disease progression in RA begins with asymptomatic autoimmune responses in cases with a genetic or environmental predisposition, that alters to arthralgia phase as autoantibodies reach the joints and subjects begin demonstrating nonspecific musculoskeletal presentations lacking any clinical symptoms of synovial inflammation. After that, patients' symptoms develop to undifferentiated arthritis (UA)/idiopathic arthritis (IA) whenever the subjects progress to clinical synovitis systemic comorbidities affecting the vasculature, metabolism, and bone, and eventually with augmented immune cell infiltration, IA/UA patients progress to clinically classifiable RA. RA is mainly correlated with different immune cells and each of them contributes variously to the pathogenesis of the disease. The pathogenesis of RA is altered by the contribution of both T and B cells in an autoimmune irregularity. Modulation of the immune responses occurs through regulatory and inhibitory molecules that control activation of the adaptive system as well as immune hemostasis. To confine the exorbitant T cell-associated inflammatory reactions, the immune system provides a system of inhibitory feedbacks, collectively named immune checkpoints. In this review, we aimed to discuss about inhibitory members of immune checkpoint molecules, including programmed cell death 1 (PD-1)/PD-L1, cytotoxic-T-lymphocyte-antigen-4, lymphocyte activation gene-3, T cell immunoglobulin-3, V-domain Ig suppressor of T cell activation, B- and T-lymphocyte attenuator, and T cell immunoglobulin and ITIM domain and their role in RA.
Collapse
Affiliation(s)
- Maryam Hemmatzadeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elham Ahangar Parvin
- Department of Biology, Faculty of Natural Science, University of Tabriz, Tabriz, Iran
| | - Hamed Mohammadi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran.,Department of Immunology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Gholamreza Azizi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Navid Shomali
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Research Center for Integrative Medicine in Aging, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
13
|
Mohammadi P, Hesari M, Chalabi M, Salari F, Khademi F. An overview of immune checkpoint therapy in autoimmune diseases. Int Immunopharmacol 2022; 107:108647. [DOI: 10.1016/j.intimp.2022.108647] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 02/17/2022] [Accepted: 02/20/2022] [Indexed: 02/06/2023]
|
14
|
Gertel S, Polachek A, Elkayam O, Furer V. Lymphocyte activation gene-3 (LAG-3) regulatory T cells: An evolving biomarker for treatment response in autoimmune diseases. Autoimmun Rev 2022; 21:103085. [PMID: 35341974 DOI: 10.1016/j.autrev.2022.103085] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 02/23/2022] [Accepted: 03/23/2022] [Indexed: 11/25/2022]
Abstract
Regulatory T cells (Tregs) comprise a CD4+CD25+Foxp3+ T cell subset for maintaining immune tolerance, and their deficits and/or dysfunction are observed in autoimmune diseases. The lymphocyte activation gene 3 (LAG-3, also known as CD223), which is an immunoglobulin superfamily member expressed on peripheral immune cells, is recognized as an inhibitory regulator of Tregs. LAG-3+ T cells represent a novel protective Tregs subset that produces interleukin-10. Alterations in LAG-3+ Tregs have been reported in several autoimmune diseases, suggesting their potential pathogenic role. Recent studies have indicated that LAG-3+ Tregs may be associated not only with immunopathology but also with response to therapy in several autoimmune and autoinflammatory diseases, such as rheumatoid arthritis, psoriasis, psoriatic arthritis and others. We present a review of Tregs phenotypes and functions, with a focus on LAG-3+ Tregs, and discuss their potential role as biomarkers for treatment response in autoimmune diseases.
Collapse
Affiliation(s)
- Smadar Gertel
- Department of Rheumatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel, affiliated to the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Ari Polachek
- Department of Rheumatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel, affiliated to the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ori Elkayam
- Department of Rheumatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel, affiliated to the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Victoria Furer
- Department of Rheumatology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel, affiliated to the Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
15
|
Gertel S, Polachek A, Furer V, Levartovsky D, Elkayam O. CD4 + LAG-3 + T cells are decreased in active psoriatic arthritis patients and their restoration in vitro is mediated by TNF inhibitors. Clin Exp Immunol 2021; 206:173-183. [PMID: 34309834 PMCID: PMC8506121 DOI: 10.1111/cei.13646] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 07/04/2021] [Accepted: 07/19/2021] [Indexed: 12/25/2022] Open
Abstract
Psoriatic arthritis (PsA) is a chronic inflammatory disease associated with T cell dysregulation. The lymphocyte-activation gene (LAG)-3 is one of the regulatory receptors expressed on T cells in a soluble form. LAG-3 expression on T cells was analyzed in vitro in PsA patients with minimal disease activity (MDA), active disease (non-MDA) and healthy controls. In cultured in-vitro peripheral blood mononuclear cells (PBMCs), LAG-3 expression on CD4+ T cells was similar in both MDA PsA patients (7.5 ± 0.9) (n = 14) and healthy controls (7.8 ± 0.6) (n = 15), but significantly lower in non-MDA PsA patients (3.1 ± 0.3) (n = 13) (p < 0.0001). An inverse correlation between PsA clinical disease activity and %CD4+ LAG-3+ T cells in vitro was observed (composite psoriatic disease activity index r = -0.47, p < 0.02 and psoriatic arthritis disease activity score, r = -0.51, p < 0.008). In-vitro co-culture of CD4+ T cells with anti-tumor necrosis factor (TNF) or anti-interleukin (IL)-17A had no effect on LAG-3+ expression in MDA PsA patients and healthy controls. In non-MDA patients, anti-TNF, but not anti-IL-17A, restored the %CD4+ LAG-3+ T cells (7.9 ± 0.9 and 3.2 ± 0.4, respectively) (p < 0.0004). Lower soluble LAG-3 levels were found in sera of naive to biological PsA patients (n = 39) compared to healthy controls (n = 35) (p < 0.03). Impaired LAG-3 on CD4+ T cells may reflect active PsA disease state. Anti-TNFs have potency to up-regulate the CD4+ LAG-3+ T cells in vitro.
Collapse
Affiliation(s)
- Smadar Gertel
- Department of RheumatologyTel Aviv Sourasky Medical CenterTel AvivIsrael
- Sackler Faculty of MedicineTel Aviv UniversityTel AvivIsrael
| | - Ari Polachek
- Department of RheumatologyTel Aviv Sourasky Medical CenterTel AvivIsrael
- Sackler Faculty of MedicineTel Aviv UniversityTel AvivIsrael
| | - Victoria Furer
- Department of RheumatologyTel Aviv Sourasky Medical CenterTel AvivIsrael
- Sackler Faculty of MedicineTel Aviv UniversityTel AvivIsrael
| | - David Levartovsky
- Department of RheumatologyTel Aviv Sourasky Medical CenterTel AvivIsrael
- Sackler Faculty of MedicineTel Aviv UniversityTel AvivIsrael
| | - Ori Elkayam
- Department of RheumatologyTel Aviv Sourasky Medical CenterTel AvivIsrael
- Sackler Faculty of MedicineTel Aviv UniversityTel AvivIsrael
| |
Collapse
|
16
|
Li X, Xu H, Huang J, Luo D, Lv S, Lu X, Xiao C. Dysfunctions, Molecular Mechanisms, and Therapeutic Strategies of Regulatory T Cells in Rheumatoid Arthritis. Front Pharmacol 2021; 12:716081. [PMID: 34512345 PMCID: PMC8428974 DOI: 10.3389/fphar.2021.716081] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 07/19/2021] [Indexed: 12/20/2022] Open
Abstract
Regulatory T cells (Tregs) represent a distinct subpopulation of CD4+ T lymphocytes that promote immune tolerance and maintain immune system homeostasis. The dysfunction of Tregs is tightly associated with rheumatoid arthritis (RA). Although the complex pathogenic processes of RA remain unclear, studies on Tregs in RA have achieved substantial progress not only in fundamental research but also in clinical application. This review discusses the current knowledge of the characterizations, functions, and molecular mechanisms of Tregs in the pathogenesis of RA, and potential therapies for these disorders are also involved.
Collapse
Affiliation(s)
- Xiaoya Li
- The Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences/Peking Union Medical College, Beijing, China.,Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Huihui Xu
- Beijing Key Laboratory of Research of Chinese Medicine on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jing Huang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Dan Luo
- Department of Ophthalmology, Traditional Chinese Medicine Hospital of Changping District, Beijing, China
| | - Shuang Lv
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China
| | - Xiangchen Lu
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China.,School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Cheng Xiao
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, China.,Department of Emergency, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
17
|
Carvajal Alegria G, Cornec D, Saraux A, Devauchelle-Pensec V, Jamin C, Hillion S, Pers JO, Pochard P. Abatacept Promotes Regulatory B Cell Functions, Enhancing Their Ability to Reduce the Th1 Response in Rheumatoid Arthritis Patients through the Production of IL-10 and TGF-β. THE JOURNAL OF IMMUNOLOGY 2021; 207:470-482. [PMID: 34244295 DOI: 10.4049/jimmunol.2000455] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 05/14/2021] [Indexed: 11/19/2022]
Abstract
Abatacept mimics natural CD152 and competes with CD28 for binding to CD80/CD86 on APC, such as B cells, thereby preventing T cell activation. However, its potential impact on B cells has not been identified. The aim of this study was to assess whether abatacept can potentiate the immunoregulatory properties of B cells in vitro and in patients with rheumatoid arthritis (RA). T and B cells from healthy controls were purified. The suppressor properties of B cells in the presence of abatacept or control IgG1 were evaluated based on the ability of these cells to inhibit the polyclonal expansion (anti-CD3/CD28 stimulation) of T cells or their differentiation into Th1 or Th17 cells. Similar analyses were also performed with cells from RA patients before and 3 mo after abatacept initiation. Abatacept significantly potentiated regulatory B cell regulatory functions by enhancing their ability to produce IL-10 and TGF-β, resulting in the increased generation of regulatory T cells and limited T cell proliferation and differentiation into Th1 and Th17 cells. Interestingly, B cells isolated from patients that received a 3-mo treatment with abatacept had an increased ability to reduce T cell functions, confirming the above observations. Abatacept binding to CD80/CD86 induces and promotes regulatory B cell functions by enhancing the ability of these cells to produce IL-10 and TGF-β in vitro and in RA patients.
Collapse
Affiliation(s)
- Guillermo Carvajal Alegria
- UMR 1227 "Lymphocytes B et Autoimmunité," Université de Brest, INSERM, LabEx IGO, Brest, France; and CHU de Brest, Brest, France
| | - Divi Cornec
- UMR 1227 "Lymphocytes B et Autoimmunité," Université de Brest, INSERM, LabEx IGO, Brest, France; and CHU de Brest, Brest, France
| | - Alain Saraux
- UMR 1227 "Lymphocytes B et Autoimmunité," Université de Brest, INSERM, LabEx IGO, Brest, France; and CHU de Brest, Brest, France
| | - Valérie Devauchelle-Pensec
- UMR 1227 "Lymphocytes B et Autoimmunité," Université de Brest, INSERM, LabEx IGO, Brest, France; and CHU de Brest, Brest, France
| | - Christophe Jamin
- UMR 1227 "Lymphocytes B et Autoimmunité," Université de Brest, INSERM, LabEx IGO, Brest, France; and CHU de Brest, Brest, France
| | - Sophie Hillion
- UMR 1227 "Lymphocytes B et Autoimmunité," Université de Brest, INSERM, LabEx IGO, Brest, France; and CHU de Brest, Brest, France
| | - Jacques-Olivier Pers
- UMR 1227 "Lymphocytes B et Autoimmunité," Université de Brest, INSERM, LabEx IGO, Brest, France; and CHU de Brest, Brest, France
| | - Pierre Pochard
- UMR 1227 "Lymphocytes B et Autoimmunité," Université de Brest, INSERM, LabEx IGO, Brest, France; and CHU de Brest, Brest, France
| |
Collapse
|
18
|
Pan L, Wang J, Liu J, Guo L, Yang S. Deficiency in the frequency and function of Tr1 cells in IgAV and the possible role of IL-27. Rheumatology (Oxford) 2021; 60:3432-3442. [PMID: 33280050 PMCID: PMC8516516 DOI: 10.1093/rheumatology/keaa752] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 10/01/2020] [Indexed: 12/14/2022] Open
Abstract
Objective Type 1 regulatory T (Tr1) cells are involved in the pathogenesis of numerous immune-mediated diseases. However, little is known about whether and how Tr1 cells affect the development of IgA vasculitis (IgAV). We aimed to investigate this question in IgAV patients. Methods . Tr1 cells in peripheral blood and kidney tissue of IgAV patients were analysed by multi-parametric flow cytometry and immunofluorescence techniques. An in vitro assay of suppression of T cell proliferation and cytokine release was performed to evaluate the function of Tr1 cells. Real-time PCR and cell stimulation in vitro were used to explore the roles of IL-27 and early growth response gene 2 (EGR2). Results The frequency of Tr1 cells was decreased in peripheral blood but increased in kidney tissue from IgAV patients. A defective suppressive function of Tr1 cells in IgAV was observed. The frequency of Tr1 cells and the cytokines secreted by them were up-regulated in the presence of recombinant IL-27 in vitro. Moreover, IL-27 also increased the expression of EGR2. Furthermore, lower frequency of Tr1 cells during remission had a higher recurrence rate. Conclusion Tr1 cells are involved in the pathogenesis of IgAV. The low IL-27 in IgAV is responsible for impaired frequency and function of Tr1 cells, and EGR2 may be the specific transcription factor involved in the progression. Tr1 may be a risk factor for IgAV recurrence.
Collapse
Affiliation(s)
- Lu Pan
- Department of Pediatric Rheumatology and Allergy, The First Hospital of Jilin University, Changchun, China
| | - Jinghua Wang
- Department of Pediatric Rheumatology and Allergy, The First Hospital of Jilin University, Changchun, China
| | - Jinxiang Liu
- Department of Pediatric Rheumatology and Allergy, The First Hospital of Jilin University, Changchun, China
| | - Lishuang Guo
- Department of Pediatric Rheumatology and Allergy, The First Hospital of Jilin University, Changchun, China
| | - Sirui Yang
- Department of Pediatric Rheumatology and Allergy, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
19
|
Alenazy MF, Saheb Sharif-Askari F, Omair MA, El-Wetidy MS, Omair MA, Mitwalli H, Al-Muhsen S, Al-Masri A, Hamid Q, Halwani R. Abatacept enhances blood regulatory B cells of rheumatoid arthritis patients to a level that associates with disease remittance. Sci Rep 2021; 11:5629. [PMID: 33707483 PMCID: PMC7952390 DOI: 10.1038/s41598-021-83615-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Accepted: 01/29/2021] [Indexed: 12/19/2022] Open
Abstract
Abatacept, an inhibitor of CD28 mediated T-cell activation, has been shown to be effective in controlling inflammation during rheumatoid arthritis (RA). However, its effects on immune regulatory B and T cells (Bregs and Tregs) has not been fully explored. Thirty-one RA patients treated with abatacept for ≥ 6 months along with 31 RA patients treated with other modalities as well as 30 healthy controls were recruited. Of these 62 RA patient, 49 (79%) were females with a mean age of 54 ± 12 years and disease duration of 10 ± 6 years. The blood levels of Tregs and Bregs and their production of immunosuppressive cytokines, were determined using FACS analysis and Luminex Multiplex assay. Treatment with abatacept significantly enhanced the blood level of IL-35+ IL-10+ Bregs (P = 0.0007). Their levels were higher in the blood of remitted patients (DAS28-CRP < 2.6) compared to the unremitted ones (P = 0.0173), 6 months following abatacept treatment initiation. Moreover, abatacept treatment significantly enhanced the blood levels of LAG3+ conventional and unconventional Tregs of RA patients. This increase in the blood levels of Bregs and Tregs was accompanied with an elevated serum level of IL-35 and IFN-β in abatacept-treated patients. Therefore, Abatacept efficiency to achieve remittance in RA could be attributed, in part, to its ability to enhance immune regulatory cells, especially IL-135+ IL-10+ Bregs.
Collapse
Affiliation(s)
- Maha Fahad Alenazy
- Immunology Research Lab, College of Medicine, King Saud University, Riyadh, Saudi Arabia.,Department of Physiology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | | | - Mohammed A Omair
- Rheumatology Unit, Department of Medicine, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Mohammad S El-Wetidy
- Immunology Research Lab, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Maha A Omair
- Department of Statistics and Operations Research, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Hussam Mitwalli
- Immunology Research Lab, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Saleh Al-Muhsen
- Immunology Research Lab, College of Medicine, King Saud University, Riyadh, Saudi Arabia.,Department of Pediatrics, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Abeer Al-Masri
- Department of Physiology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Qutayba Hamid
- Sharjah Institute of Medical Research, University of Sharjah, Sharjah, United Arab Emirates.,Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.,Meakins-Christie Laboratories, Research Institute of the McGill University Health Center, Montreal, QC, Canada
| | - Rabih Halwani
- Sharjah Institute of Medical Research, University of Sharjah, Sharjah, United Arab Emirates. .,Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates. .,Prince Abdullah Ben Khaled Celiac Disease Chair, College of Medicine, King Saud University, Riyadh, Saudi Arabia.
| |
Collapse
|
20
|
Greisen SR, Deleuran B. Checkpoint Molecules in Rheumatology-or the Benefits of Being Exhausted. Curr Rheumatol Rep 2021; 23:22. [PMID: 33651184 DOI: 10.1007/s11926-021-00991-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2021] [Indexed: 12/18/2022]
Abstract
PURPOSE OF REVIEW This review will focus on the most common co-inhibitory molecules, emphasizing the importance of these in relation to rheumatic disease. RECENT FINDINGS Checkpoint molecules are pivotal in determining the outcome of antigen activation. Checkpoint molecules consist of co-stimulatory and co-inhibitory molecules, where the first activates and the latter inhibits the antigen presentation process. Studies show that increased activity of co-inhibitory molecules is associated with a good prognosis in rheumatic diseases. Opposite, when cancer patients are treated with antibodies blocking the inhibitory pathways, autoimmune diseases, including arthritis, develop as immune-related adverse events (IrAE). This emphasizes the importance of these pathways in autoimmune disease. Co-inhibitory molecules are becoming increasingly interesting as future treatment targets in rheumatic conditions. Treatments with antibodies blocking these pathways result in IrAE, often manifesting as autoimmune rheumatic diseases. Therefore, a need to get acquainted with these molecules is growing so we can cope with future challenges in rheumatic diseases.
Collapse
Affiliation(s)
- Stinne Ravn Greisen
- Department of Biomedicine, Aarhus University, Skou-building, C.F. Møllers Alle 6, DK-8000, Aarhus C, Denmark. .,Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark.
| | - Bent Deleuran
- Department of Biomedicine, Aarhus University, Skou-building, C.F. Møllers Alle 6, DK-8000, Aarhus C, Denmark.,Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
21
|
Novel human immunomodulatory T cell receptors and their double-edged potential in autoimmunity, cardiovascular disease and cancer. Cell Mol Immunol 2020; 18:919-935. [PMID: 33235388 DOI: 10.1038/s41423-020-00586-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 10/28/2020] [Indexed: 12/15/2022] Open
Abstract
In the last decade, approaches based on T cells and their immunomodulatory receptors have emerged as a solid improvement in treatments for various types of cancer. However, the roles of these molecules in the therapeutic context of autoimmune and cardiovascular diseases are still relatively unexplored. Here, we review the best known and most commonly used immunomodulatory T cell receptors in clinical practice (PD-1 and CTLA-4), along with the rest of the receptors with known functions in animal models, which have great potential as modulators in human pathologies in the medium term. Among these other receptors is the receptor CD69, which has recently been described to be expressed in mouse and human T cells in autoimmune and cardiovascular diseases and cancer. However, inhibition of these receptors individually or in combination by drugs or monoclonal antibodies generates a loss of immunological tolerance and can trigger multiple autoimmune disorders in different organs and immune-related adverse effects. In the coming decades, knowledge on the functions of different immunomodulatory receptors will be pivotal for the development of new and better therapies with less harmful side effects. In this review, we discuss the roles of these receptors in the control of immunity from a perspective focused on therapeutic potential in not only cancer but also autoimmune diseases, such as systemic lupus erythematosus, autoimmune diabetes and rheumatoid arthritis, and cardiovascular diseases, such as atherosclerosis, acute myocardial infarction, and myocarditis.
Collapse
|
22
|
Ding J, Liu M, Xuan Z, Liu ML, Wang N, Jia X. The Protective Effects of the Ethyl Acetate Part of Er MiaoSan on Adjuvant Arthritis Rats by Regulating the Function of Bone Marrow-Derived Dendritic Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2020; 2020:8791657. [PMID: 39295892 PMCID: PMC11410441 DOI: 10.1155/2020/8791657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 10/05/2020] [Accepted: 10/28/2020] [Indexed: 09/21/2024]
Abstract
Aims The aim of this study was to evaluate the protective effects of Er Miao San (EMS) and the regulative function of bone marrow-derived dendritic cells (BMDCs) on adjuvant arthritis (AA) in rats. Methods The ethyl acetate part of EMS (3 g/kg, 1.5 g/kg, and 0.75 g/kg) was orally administered from day 15 after immunization to day 29. The polyarthritis index and paw swelling were measured, the ankle joint pathological changes were observed using hematoxylin-eosin (HE) staining, and the spleen and thymus index were determined. Moreover, T and B cell proliferation were determined using the CCK-8 assay. The expression of BMDC surface costimulatory molecules and inflammatory factors were determined using flow cytometry and ELISA kits, respectively. Results Compared with the AA model rats, the ethyl acetate fraction of EMS obviously reduced paw swelling (from 1.0 to 0.7) and the polyarthritis index (from 12 to 9) (P < 0.01) and improved the severity of histopathology (P < 0.01). The treatment using ethyl acetate fraction of EMS significantly reduced the spleen and thymus index (P < 0.01) and inhibited T and B cell proliferation (P < 0.01). Moreover, EMS significantly modulated the expression of surface costimulatory molecules in BMDCs, including CD40, CD80, CD86, and major histocompatibility complex class II (MHC-II) (P < 0.01). The results also showed that the ethyl acetate part of EMS significant inhibited the levels of proinflammatory cytokines interleukin- (IL-) 23 tumor necrosis factor- (TNF-) α and inflammatory factor prostaglandin (PG) E2 in the supernatant of BMDCs. However, the level of anti-inflammatory cytokine IL-10 was significantly increased (P < 0.01). Conclusion These results suggest that the ethyl acetate part of EMS has better protective effects on AA rats, probably by regulating the function of BMDCs and modulating the balance of cytokines.
Collapse
Affiliation(s)
- Jiemin Ding
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, Anhui 230012, China
- Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei 230012, China
| | - Min Liu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, Anhui 230012, China
- Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei 230012, China
| | - Zihua Xuan
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, Anhui 230012, China
- Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei 230012, China
| | - Meng Li Liu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, Anhui 230012, China
- Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei 230012, China
| | - Ning Wang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, Anhui 230012, China
- Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei 230012, China
| | - Xiaoyi Jia
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
- Anhui Province Key Laboratory of Chinese Medicinal Formula, Hefei, Anhui 230012, China
- Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Hefei 230012, China
| |
Collapse
|
23
|
Chauhan RK, Sharma PK, Srivastava S. Role of signaling pathway in biological cause of Rheumatoid arthritis. Curr Drug Res Rev 2020; 13:130-139. [PMID: 33172384 DOI: 10.2174/2589977512999201109215004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 07/14/2020] [Accepted: 09/03/2020] [Indexed: 11/22/2022]
Abstract
Rheumatoid Arthritis is a chronic progressive inflammatory auto-immune disease in which the immune system of the body attacks its cartilage and joints lining. It not only affects synovial joints but also many other sites including heart, blood vessels, and skins. It is more common in females than in males. The exact cause of rheumatoid arthritis is not well established but the hypothesis reported in the literature is that in the development stage of the disease, both genetics and environmental factors can play an inciting role. Along with these factors alteration in the normal physiology of enzymatic action, acts as a trigger to develop this condition. Numerous signaling pathways involved in the pathogenesis of Rheumatoid Arthritis involves activation of mitogen-activated protein kinase, kinases Janus family, P-38 Mitogen-Activated Protein Kinase, Nuclear Factor-kappa B. Interleukin-1 to play a proinflammatory cytokine that plays an important role in inflammation in RA. These are also associated with an increase in neutrophil, macrophage and lymphocytic chemotaxis, mast cell degranulation, activation, maturation and survival of T-cells and B-cells activated. These signaling pathways also show that p38α downregulation in myeloid cells exacerbates the severity of symptoms of arthritis. Thus, present review carters about the detail of different signaling pathways and their role in rheumatoid arthritis.
Collapse
Affiliation(s)
- Rakesh Kumar Chauhan
- Department of Pharmacy, School of Medical and Allied Science, Galgotias University, Plot N. 2, Sector 17- A, Yamuna Expressway, Gautam Buddha Nagar, Greater Noida, Uttar Pradesh 201306,. India
| | - Pramod Kumar Sharma
- Department of Pharmacy, School of Medical and Allied Science, Galgotias University, Plot N. 2, Sector 17- A, Yamuna Expressway, Gautam Buddha Nagar, Greater Noida, Uttar Pradesh 201306,. India
| | - Shikha Srivastava
- Department of Pharmacy, School of Medical and Allied Science, Galgotias University, Plot N. 2, Sector 17- A, Yamuna Expressway, Gautam Buddha Nagar, Greater Noida, Uttar Pradesh 201306,. India
| |
Collapse
|
24
|
Liu S, Guo Y, Lu L, Lu J, Ke M, Xu T, Lu Y, Chen W, Wang J, Kong D, Shen Q, Zhu Y, Tan W, Ji W, Zhou W. Fibrinogen-Like Protein 1 Is a Novel Biomarker for Predicting Disease Activity and Prognosis of Rheumatoid Arthritis. Front Immunol 2020; 11:579228. [PMID: 33123164 PMCID: PMC7574527 DOI: 10.3389/fimmu.2020.579228] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 08/20/2020] [Indexed: 12/17/2022] Open
Abstract
Rheumatoid arthritis (RA), afflicting over 1% of the population, is an inflammatory joint disease leading to cartilage damage and ultimately impaired joint function. Disease-modifying anti-rheumatic drugs are considered as the first-line treatment to inhibit the progression of RA, and the treatment depends on the disease status assessment. The disease activity score 28 as clinical gold standard is extensively used for RA assessment, but it has the limitations of delayed assessment and the need for specialized expertise. It is necessary to discover biomarkers that can precisely monitor disease activity, and provide optimized treatment for RA patients. A total of 1,244 participants from two independent centers were divided into five cohorts. Cohorts 1–4 constituted sera samples of moderate to high active RA, low active RA, RA in remission and healthy subjects. Cohort 5 consisted of sera of RA, osteoarthritis (OA), ankylosing spondylitis (AS), systemic lupus erythematosus (SLE), primary Sjogren's syndrome (pSS) and healthy subjects. Biomarkers were found from cohorts 1–2 (screening sets), cohort 3 (discovery and external validation sets), cohort 4 (drug intervention set) and cohort 5 (biomarker-specific evaluation set). We found 68 upregulated and 74 downregulated proteins by TMT-labeled proteomics in cohort 1, and fibrinogen-like protein 1 (FGL1) had the highest area under the receiver operating characteristic curve (AUC) values in cohort 2. In cohort 3, in cross-comparison among moderate/high active RA, low active RA, RA in remission and healthy subjects, FGL1 had AUC values of approximately 0.9000 and predictive values of 90%. Additionally, FGL1 had a predictive value of 91.46% for moderate/high active RA vs. remission/low active RA and 80.77% for RA in remission vs. low active RA in cohort 4. Importantly, FGL1 levels had no significant difference in OA and AS compared with healthy persons. The concentrations in SLE and pSS were improved, but approximately 3-fold lower than that in active RA in cohort 5. In summary, FGL1 is a novel and specific biomarker that could be clinically useful for predicting progression of RA.
Collapse
Affiliation(s)
- Shijia Liu
- Department of Rheumatology and Immunology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yunke Guo
- Department of Rheumatology and Immunology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Lu Lu
- Department of Rheumatology and Immunology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jiawei Lu
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Mengying Ke
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Tingting Xu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yan Lu
- Department of Rheumatology and Immunology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Wenjun Chen
- Department of Rheumatology and Immunology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jue Wang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Deshun Kong
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Qiuxiang Shen
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Youjuan Zhu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - WenFeng Tan
- Department of Rheumatology and Immunology, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Wei Ji
- Department of Rheumatology and Immunology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Wei Zhou
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
25
|
Symonds AL, Zheng W, Miao T, Wang H, Wang T, Kiome R, Hou X, Li S, Wang P. Egr2 and 3 control inflammation, but maintain homeostasis, of PD-1 high memory phenotype CD4 T cells. Life Sci Alliance 2020; 3:3/9/e202000766. [PMID: 32709717 PMCID: PMC7391068 DOI: 10.26508/lsa.202000766] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 07/09/2020] [Accepted: 07/10/2020] [Indexed: 01/13/2023] Open
Abstract
PD-1high memory CD4 T cells are pathogenic in autoimmune disease; here they show their expression of Egr2 is defective in rheumatoid arthritis and Egr2 & 3 control their inflammation and homeostasis. The transcription factors Egr2 and 3 are essential for controlling inflammatory autoimmune responses of memory phenotype (MP) CD4 T cells. However, the mechanism is still unclear. We have now found that the Egr2+ subset (PD-1high MP) of MP CD4 T cells expresses high levels of checkpoint molecules (PD-1 and Lag3) and also markers of effector T cells (CXCR3 and ICAM-1). Egr2/3 are not required for PD-1high MP CD4 cell development but mediate a unique transcriptional programme that effectively controls their inflammatory responses, while promoting homeostatic proliferation and adaptive responses. Egr2 negative PD-1high MP CD4 T cells are impaired in homeostatic proliferation and adaptive responses against viral infection but display inflammatory responses to innate stimulation such as IL-12. PD-1high MP CD4 T cells have recently been implicated in rheumatoid arthritis pathogenesis, and we have now found that Egr2 expression is reduced in PD-1high MP CD4 T cells from patients with active rheumatoid arthritis compared with healthy controls. These findings demonstrate that Egr2/3 control the inflammatory responses of PD-1high MP CD4 T cells and maintain their adaptive immune fitness.
Collapse
Affiliation(s)
- Alistair Lj Symonds
- The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Wei Zheng
- Division of Rheumatology, Dong Fang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Tizong Miao
- The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Haiyu Wang
- Division of Rheumatology, Dong Fang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - TieShang Wang
- Division of Rheumatology, Dong Fang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Ruth Kiome
- Bioscience, Brunel University, Uxbridge, UK
| | - Xiujuan Hou
- Division of Rheumatology, Dong Fang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Suling Li
- Bioscience, Brunel University, Uxbridge, UK
| | - Ping Wang
- The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
26
|
Mohammadzadeh A. Co-inhibitory receptors, transcription factors and tolerance. Int Immunopharmacol 2020; 84:106572. [DOI: 10.1016/j.intimp.2020.106572] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/20/2020] [Accepted: 05/04/2020] [Indexed: 12/23/2022]
|
27
|
Hu S, Liu X, Li T, Li Z, Hu F. LAG3 (CD223) and autoimmunity: Emerging evidence. J Autoimmun 2020; 112:102504. [PMID: 32576412 DOI: 10.1016/j.jaut.2020.102504] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/07/2020] [Accepted: 06/10/2020] [Indexed: 12/31/2022]
Abstract
Immune checkpoint molecules play pivotal roles in maintaining the immune homeostasis. Targeting these molecules, such as the classical Cytotoxic T-Lymphocyte Antigen 4 (CTLA4) and Programmed Cell Death Protein 1 (PD1), achieves great success in treating cancers. However, not all the patients respond well. This urges the immunologists to identify novel immune checkpoint molecules. Lymphocyte activation gene-3 (LAG3; CD223) is a newly identified inhibitory receptor. It is expressed on a variety of immune cells, including CD4+ T cells, CD8+ T cells, Tregs, B cells, and NK cells. Its unique intracellular domains, signaling patterns as well as the striking synergy observed in its targeted therapy with anti-PD1 indicate the important role of LAG3 in maintaining immune tolerance. Currently, a variety of agents targeting LAG3 are in clinical trials, revealing great perspectives in the future immunotherapy. In this review, we briefly summarize the studies on LAG3, including its structure, isoforms, ligands, signaling, function, roles in multiple diseases, as well as the latest targeted therapeutic advances, with particular concern on the potential association of LAG3 with autoimmune diseases.
Collapse
Affiliation(s)
- Suiyuan Hu
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Xu Liu
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Tianding Li
- Software Center, Bank of China, Beijing, China
| | - Zhanguo Li
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China; State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Fanlei Hu
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China; State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China.
| |
Collapse
|
28
|
CTLA4-Ig Abatacept Ameliorates Proteinuria by Regulating Circulating Treg/IL-17 in Adriamycin-Induced Nephropathy Rats. BIOMED RESEARCH INTERNATIONAL 2020; 2020:2347827. [PMID: 32420329 PMCID: PMC7201454 DOI: 10.1155/2020/2347827] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 02/09/2020] [Accepted: 03/20/2020] [Indexed: 12/30/2022]
Abstract
Objective This study is aimed at investigating the efficacy of CTLA4-Ig abatacept in normalizing proteinuria and its possible mechanism in adriamycin-induced nephropathy (AIN) rats. Methods A total of 32 healthy male Sprague-Dawley rats were randomly divided into a normal group, an AIN group, an abatacept group, and a prednisone group. Adriamycin (6.5 mg/kg) was injected once via the tail vein of rats to induce nephrotic syndrome. After adriamycin treatment, the abatacept group rats were given abatacept (0.5 mg/kg) once by intraperitoneal injection on day 14. In addition, the prednisone group rats were given prednisone (12.5 mg/kg) daily consecutively by gavage from day 14 to day 21. Blood, urine, and kidney tissue specimens were collected when sacrificed on day 21. The 24-hour urinary protein, serum albumin, cholesterol, creatinine, and urea nitrogen were then detected. An enzyme-linked immunosorbent assay was used to determine the level of urine CD80 and serum IL-17. Flow cytometry was used to investigate the prevalence of circulating Treg. Hematoxylin-eosin staining and electron microscopy were used for a renal histological study. Immunofluorescence staining was performed to confirm the CD80 expression of renal tissue. Results The 24-hour urinary protein of the abatacept group was significantly lower than that of the prednisone group and the AIN group. The level of urine CD80 of the abatacept group was significantly lower than that of the AIN group. Compared with the AIN group and the prednisone group, the circulating Treg prevalence of the abatacept group was significantly higher, while the level of serum IL-17 was lower. A negative kidney staining of CD80 expression was demonstrated in each group in this study. The 24-hour urinary protein had a negative correlation with the circulating Treg prevalence and Treg/IL-17 and a positive correlation with the urine CD80 and serum IL-17. Urinary CD80 had a positive correlation with serum IL-17 and no correlation with the circulating Treg prevalence. Conclusions CTLA4-Ig abatacept can reduce proteinuria of adriamycin-induced nephropathy rats, possibly at least partially as a result of regulating circulating Treg/IL-17. CTLA4-Ig abatacept could be a promising regimen for idiopathic nephrotic syndrome.
Collapse
|
29
|
Angin M, Brignone C, Triebel F. A LAG-3-Specific Agonist Antibody for the Treatment of T Cell-Induced Autoimmune Diseases. THE JOURNAL OF IMMUNOLOGY 2020; 204:810-818. [PMID: 31907283 DOI: 10.4049/jimmunol.1900823] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 12/06/2019] [Indexed: 12/18/2022]
Abstract
T cells chronically stimulated with the same peptide tend to express exhaustion markers such as PD-1 or LAG-3. Deficiencies in the PD-1 and LAG-3 pathways have been linked to the development of autoimmune diseases. IMP761 is a LAG-3-specific humanized agonist Ab with immunosuppressive properties both in vitro and in vivo in an Ag-specific delayed-type hypersensitivity (DTH) model in the cynomolgus macaque (Macaca fascicularis). IMP761 inhibits TCR-mediated NFAT activation and Ag-induced human T cell proliferation and activation. In the DTH model, assessment of T cell infiltration and gene expression profile at the DTH biopsy site corresponds to immunosuppression of an Ag-induced T cell response. IMP761 is the first LAG-3-specific agonist product candidate, acting upstream on activated T cells, the root cause of self-Ag-specific T cell-induced autoimmune diseases.
Collapse
|
30
|
Zhang C, Jiao S, Li T, Zhao L, Chen H. Relationship between polymorphisms in -572G/C interleukin 6 promoter gene polymorphisms (rs1800796) and risk of rheumatoid arthritis: A meta-analysis. Int J Rheum Dis 2019; 23:47-54. [PMID: 31782615 DOI: 10.1111/1756-185x.13729] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 07/21/2019] [Accepted: 09/28/2019] [Indexed: 01/10/2023]
Abstract
AIMS This meta-analysis was aimed to investigate the association between -572G/C interleukin (IL)-6 gene polymorphism and occurrence risk of rheumatoid arthritis (RA). METHODS Literature search was conducted in PubMed, Embase, Springer and Google Scholar up to November 2018. The pooled odds ratios (ORs) and 95% confidence interval (CI) were calculated by Revman 5.3. RESULTS A total of six case-control studies were included in this meta-analysis. In the allele model (G vs C), homozygous gene model (GG vs CC), recessive gene model (GG vs GC + CC), and dominant gene model (GG + GC vs CC), the pooled estimate indicated there was significant association between -572G/C IL-6 gene polymorphism and risk of RA. However, no significant statistical results were found in meta-analyses of heterozygote gene models. CONCLUSIONS The -572G/C IL-6 gene polymorphism is associated with the risk of RA. The GG genotype may be the main contributor in increasing susceptibility to RA.
Collapse
Affiliation(s)
- Chunfang Zhang
- Department of Traditional Chinese Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Shuang Jiao
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Tianyu Li
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Lihua Zhao
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Huijun Chen
- Department of Cardiology, The Second Hospital Affiliated Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| |
Collapse
|
31
|
Immune checkpoint molecules. Possible future therapeutic implications in autoimmune diseases. J Autoimmun 2019; 104:102333. [DOI: 10.1016/j.jaut.2019.102333] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 08/29/2019] [Indexed: 02/07/2023]
|
32
|
Detection of Increased Relative Expression Units of Bacteroides and Prevotella, and Decreased Clostridium leptum in Stool Samples from Brazilian Rheumatoid Arthritis Patients: A Pilot Study. Microorganisms 2019; 7:microorganisms7100413. [PMID: 31581593 PMCID: PMC6843655 DOI: 10.3390/microorganisms7100413] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Accepted: 09/25/2019] [Indexed: 12/27/2022] Open
Abstract
Interactions between gut microbes and disease modifying antirheumatic drugs (DMARDs) have been proposed. The aim of the present study was to evaluate the presence of some specific bacteria in stool samples from Brazilian RA patients receiving DMARDs and correlate these data with diet, clinical parameters, and cytokines. Stool samples were used for gut bacteria evalutation by qPCR. Serum samples were used to quantify IL-4 and IL-10 by flow cytometer. Statistics were performed by Pearson chi-square, Mann–Whitney U test, and Spearman’s correlation. The study included 20 RA patients and 30 healthy controls. There were no significant differences (p > 0.05) in dietary habits between RA patients and controls. Concerning gut bacteria, we observed an increase in relative expression units (REU) of Bacteroides and Prevotella species in stool samples from patients, and a decrease in REU of Clostridium leptum when compared with healthy controls. Positive correlation between Prevotella and rheumatoid factor was detected. The IL-4 and IL-10 concentrations were increased in patients when compared with controls. We concluded that gut bacteria are different between RA patients receiving DMARDs and healthy controls. Further studies are necessary to determine the real role of gut microbes and their metabolities in clinical response to different DMARDs in RA patients.
Collapse
|
33
|
Abstract
Rheumatoid arthritis (RA) is a chronic and progressive systemic disease of the connective tissue, which is particularly manifested with destructive alterations to the joints. Inflammatory reactions in the synovium lead to the influx of peripheral inflammatory cells as well as the activation of local cells. Released growth factors, chemokines and especially cytokines play a key role in chronic inflammatory responses. In addition to the central lymphocytes, the T and B cells and their subpopulations, locally resident cells, such as neutrophils, macrophages and fibroblasts as well as cells of bone metabolism are activated by the inflammatory milieu and contribute to and drive inflammation and tissue damage. The destruction of cartilage and bone substance by local tissue cells, synovial fibroblasts and osteoclasts is characteristic for this disease. Untreated, the local inflammatory and destructive processes as well as systemic inflammatory factors lead to progressive and irreversible joint destruction. Cellular and immunological processes in RA are closely interwoven; therefore, besides the general inhibition of immunological processes, specific inhibition of central key molecules can reduce or completely stop the inflammatory destructive processes; however, a high heterogeneity can be observed among RA patients and disease progression. Therefore, an expansion of the therapeutic options is desirable as not all patients are able to equally benefit from the therapeutic treatment. It is important to characterize new molecular mechanisms, which could lead to the development of new therapeutic options. Some of the more recent insights are summarized in this overview.
Collapse
Affiliation(s)
- E Neumann
- Rheumatologie und Klinische Immunologie, Campus Kerckhoff, Justus-Liebig-Universität Gießen, Benekestr. 2-8, 61231, Bad Nauheim, Deutschland.
| | - K Frommer
- Rheumatologie und Klinische Immunologie, Campus Kerckhoff, Justus-Liebig-Universität Gießen, Benekestr. 2-8, 61231, Bad Nauheim, Deutschland
| | - M Diller
- Rheumatologie und Klinische Immunologie, Campus Kerckhoff, Justus-Liebig-Universität Gießen, Benekestr. 2-8, 61231, Bad Nauheim, Deutschland
| | - U Müller-Ladner
- Rheumatologie und Klinische Immunologie, Campus Kerckhoff, Justus-Liebig-Universität Gießen, Benekestr. 2-8, 61231, Bad Nauheim, Deutschland
| |
Collapse
|
34
|
Islam SMS, Byun HO, Choi B, Sohn S. Inhibition of CD83 Alleviates Systemic Inflammation in Herpes Simplex Virus Type 1-Induced Behçet's Disease Model Mouse. Mediators Inflamm 2019; 2019:5761392. [PMID: 31582900 PMCID: PMC6754941 DOI: 10.1155/2019/5761392] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 07/12/2019] [Accepted: 08/08/2019] [Indexed: 11/25/2022] Open
Abstract
Behçet's disease (BD) is an autoinflammatory disease that can lead to life- and sight-threating complications. Dendritic cells (DCs) are the most potent antigen-presenting cells that can regulate multiple inflammatory pathways. The objective of this study was to investigate the association of the DC stimulatory molecule CD83 with BD. Frequencies of costimulatory molecules expressing DCs in peripheral blood leukocytes (PBL) were measured by flow cytometry (FACS). The severity of symptoms in HSV-1-induced BD symptomatic mice was also assessed. Frequencies of CD83-positive cells were significantly increased in mice exhibiting BD symptoms, compared to those in asymptomatic mice. Abatacept, a CD80/86 blocker, significantly decreased the frequencies of CD83-positive cells in a time- and dose-dependent manner. BD symptomatic mice treated with Abatacept showed gradual reduction in the severity score of symptoms. Intraperitoneal injection of CD83 siRNA significantly reduced the frequencies of CD83-positive cells in PBL and peritoneal macrophages. After CD83 siRNA injection, BD symptoms of mice were improved and disease severity was decreased. Discontinuation of CD83 siRNA deteriorated symptoms while readministration of CD83 siRNA again improved BD symptoms of mice. These results clearly indicate the involvement of CD83-expressing cells in the inflammatory symptoms of BD. Therefore, CD83 might be useful as a therapeutic target for BD.
Collapse
Affiliation(s)
- S. M. Shamsul Islam
- Department of Biomedical Science, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - Hae-Ok Byun
- Department of Microbiology, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - Bunsoon Choi
- Institute for Medical Sciences, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - Seonghyang Sohn
- Department of Biomedical Science, Ajou University School of Medicine, Suwon 16499, Republic of Korea
- Department of Microbiology, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| |
Collapse
|
35
|
Del Porto F, Cifani N, Proietta M, Dezi T, Tritapepe L, Raffa S, Micaloni A, Taurino M. Lag3 + regulatory T lymphocytes in critical carotid artery stenosis. Clin Exp Med 2019; 19:463-468. [PMID: 31302842 DOI: 10.1007/s10238-019-00570-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 07/08/2019] [Indexed: 12/13/2022]
Abstract
The aim of this study was to evaluate CD25+ and Lag3+ T regulatory subpopulations in patients with critical carotid artery stenosis (CAS) and Stanford-A acute aortic dissection (AAD). CD25+ and Lag3+ were measured in 36 patients affected by CAS and 24 patients with Stanford type A AAD. Based on neurological symptoms, patients affected by CAS were further divided in 25 asymptomatic (CAS-A) and 11 symptomatic (CAS-S) subjects. Twenty-five patients with traditional cardiovascular risk factors (RF), matched for age and sex, were used as control group. Interleukin (IL)-10, IL-6 and transforming growth factor-β-levels were also measured. CD25+ T cells were significantly increased in CAS-S versus CAS-A (p > 0.05), AAD (p > 0.05) and RF (p > 0.05). Moreover, a significant increase in Lag3+ Tregs was observed in CAS e CAS-S versus AAD (p < 0.05) and RF (p < 0.05), whereas no significant difference was observed between CAS-S and CAS-A. IL-6 was higher in AAD compared to the other groups. Patients with neurological symptoms display a peculiar expansion of CD25+ T cells, strongly confirming a relationship between ischemic brain damage and this regulatory subpopulation, whereas Lag3+ Tregs early distinguish CAS from AAD and probably exert protective actions against aortic wall rupture throughout their anti-inflammatory functions.
Collapse
Affiliation(s)
- F Del Porto
- Dipartimento di Medicina Clinica e Molecolare, Facoltà di Medicina e Psicologia, Ospedale Sant'Andrea, UOC Medicina Interna, Sapienza Università di Roma, Rome, Italy.
| | - N Cifani
- Dipartimento di Medicina Clinica e Molecolare, Facoltà di Medicina e Psicologia, Ospedale Sant'Andrea, UOC Medicina Interna, Sapienza Università di Roma, Rome, Italy
| | - M Proietta
- Dipartimento di Medicina Clinica e Molecolare, Facoltà di Medicina e Psicologia, Ospedale Sant'Andrea, UOC Medicina Interna, Sapienza Università di Roma, Rome, Italy
| | - T Dezi
- Dipartimento di Medicina Clinica e Molecolare, Facoltà di Medicina e Psicologia, Ospedale Sant'Andrea, UOC Chirurgia Vascolare, Sapienza Università di Roma, Rome, Italy
| | - L Tritapepe
- Dipartimento di Scienze Anestesiologiche, Facoltà di Medicina e Odontoiatria, Medicina Critica e Terapia del Dolore, Policlinico Umberto I, Sapienza Università di Roma, Rome, Italy
| | - S Raffa
- Dipartimento di Medicina Clinica e Molecolare, Facoltà di Medicina e Psicologia, Ospedale Sant'Andrea, UOC Diagnostica Cellulare, Sapienza Università di Roma, Rome, Italy
| | - A Micaloni
- Dipartimento di Medicina Clinica e Molecolare, Facoltà di Medicina e Psicologia, Ospedale Sant'Andrea, UOC Diagnostica Cellulare, Sapienza Università di Roma, Rome, Italy
| | - M Taurino
- Dipartimento di Medicina Clinica e Molecolare, Facoltà di Medicina e Psicologia, Ospedale Sant'Andrea, UOC Chirurgia Vascolare, Sapienza Università di Roma, Rome, Italy
| |
Collapse
|
36
|
Kato R, Sumitomo S, Tsuchida Y, Tsuchiya H, Nakachi S, Sakurai K, Hanata N, Nagafuchi Y, Kubo K, Tateishi S, Kanda H, Okamura T, Yamamoto K, Fujio K. CD4 +CD25 +LAG3 + T Cells With a Feature of Th17 Cells Associated With Systemic Lupus Erythematosus Disease Activity. Front Immunol 2019; 10:1619. [PMID: 31354747 PMCID: PMC6640175 DOI: 10.3389/fimmu.2019.01619] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Accepted: 06/28/2019] [Indexed: 12/14/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease that involves multiple immune cell subsets. We analyzed immune cell subsets in human peripheral blood mononuclear cells (PBMC) in order to identify the cells that are significantly associated with SLE disease activity and treatment. The frequencies of various subsets of CD4+ T cells, B cells, monocytes and NK cells in PBMC were assessed in 30 healthy controls (HC), 30 rheumatoid arthritis (RA) patients and 26 SLE patients using flow cytometry. The correlations between subset frequencies in SLE and clinical traits including Systemic Lupus Erythematosus Disease Activity Index (SLEDAI) were examined. Changes in subset frequencies after the treatment in SLE patients were investigated. We focused on CD25+LAG3+ T cells and investigated their characteristics, including cytokine secretion, mRNA expression and suppression capacity. We assessed correlations between CD25+LAG3+ T cells and SLEDAI by Spearman's rank correlation coefficient. CD25+LAG3+ T cells were significantly increased in SLE whereas there were few in RA and HC groups. CD25+LAG3+ T cell frequencies were significantly correlated with SLEDAI and were increased in patients with a high SLEDAI score (> 10). CD25+LAG3+ T cells produced both IL-17 and FOXP3, expressed mRNA of both FOXP3 and RORC and lacked suppressive capacity. CD25+LAG3+ T cells were associated with disease activity of SLE. CD25+LAG3+ T cells had features of both CD25+FOXP3+ regulatory T cells (CD25+ Treg) and Th17. CD25+LAG3+ T cells could be associated with the inflammatory pathophysiology of SLE.
Collapse
Affiliation(s)
- Rika Kato
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shuji Sumitomo
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yumi Tsuchida
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Haruka Tsuchiya
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shinichiro Nakachi
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Keiichi Sakurai
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Norio Hanata
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yasuo Nagafuchi
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kanae Kubo
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shoko Tateishi
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Immunotherapy Management, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroko Kanda
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Department of Immunotherapy Management, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tomohisa Okamura
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Max Planck-The University of Tokyo Center for Integrative Inflammology, The University of Tokyo, Tokyo, Japan
| | - Kazuhiko Yamamoto
- Center for Integrative Medical Sciences, The Institute of Physical and Chemical Research, Yokohama, Japan
| | - Keishi Fujio
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
37
|
Alopecia areata patients show deficiency of FOXP3+CD39+ T regulatory cells and clonotypic restriction of Treg TCRβ-chain, which highlights the immunopathological aspect of the disease. PLoS One 2019; 14:e0210308. [PMID: 31277078 PMCID: PMC6611701 DOI: 10.1371/journal.pone.0210308] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 05/28/2019] [Indexed: 12/29/2022] Open
Abstract
Alopecia areata (AA) is a hair loss disorder resulting from an autoimmune reaction against hair follicles. T-helper 1 cells are a major contributor to this disorder, but little is known about the role of T-regulatory cells (Tregs) in AA. Here, we analysed the distribution of circulating Treg subsets in twenty AA patients with active hair loss and fifteen healthy subjects by flow cytometry. The Treg suppressor HLA-DR+ subpopulation was significantly reduced in the patients (P<0.001) and there were significantly fewer cells expressing CD39 among the CD4+CD25+Foxp3+ Treg subpopulation in patients (P = 0.001). FOXP3 CD39 Treg cells were also reduced in hair follicles; by 75% in non-lesional skin and 90% in lesional skin, when compared to control healthy skin. To further characterise Treg cells in AA; Tregs (CD4+CD25+FOXP3+) were investigated for their TCRβ sequence. PCR products analysed by Next Generation Sequencing techniques, showed that all frequent public clonotypes in AA Tregs were also present in controls at relatively similar frequencies, excepting two public clonotypes: CATSRDEGGLDEKLFF (V15 D1 J1-4) and CASRDGTGPSNYGYTF (V2 D1 J1-2), which were exclusively present in controls. This suggests that these Treg clonotypes may have a protective effect and that they may be an exciting subject for future therapeutic applications.
Collapse
|
38
|
Jia X, Zhai T, Wang B, Yao Q, Li Q, Mu K, Zhang JA. Decreased number and impaired function of type 1 regulatory T cells in autoimmune diseases. J Cell Physiol 2019; 234:12442-12450. [PMID: 30666652 DOI: 10.1002/jcp.28092] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 12/07/2018] [Indexed: 12/27/2022]
Abstract
Type 1 regulatory T (Tr1) cell is a special type of T regulatory cells with surface molecular markers such as lymphocyte-activation gene 3 and CD49b. A key property of Tr1 cells is the capability to produce high-level interleukin 10 (IL-10) upon activation, in a FOXP3-independent manner. The immunosuppressive function of IL-10 producing Tr1 cells has been extensively studied for many years. Autoimmune diseases (AIDs) are conditions in which the immune system breaks down and starts to attack the body. AIDs include inflammatory bowel disease, rheumatoid arthritis, multiple sclerosis (MS), type 1 diabetes mellitus, Greaves' disease, and so forth. In recent years, more and more studies have documented that the number of Tr1 cells is decreased and the function is inhibited in a variety of AIDs, among which MS is the most widely studied. The protocol for engineering Tr1 cell therapy has been established and is gradually being used in clinical practice in recent years. Tr1 cell therapy has been proven to be safe and effective, but it is mainly involved in myeloid leukemia, graft versus host disease currently. Its therapeutic role in AIDs still needs to be further explored. In this study, we will summarize the research advances of Tr1 cells in AIDs, which will provide useful information for treating AIDs through Tr1 cell therapy in the future.
Collapse
Affiliation(s)
- Xi Jia
- Department of Endocrinology, Jinshan Hospital of Fudan University, Shanghai, China
| | - Tianyu Zhai
- Department of Endocrinology, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Bing Wang
- Department of Endocrinology, Jinshan Hospital of Fudan University, Shanghai, China
| | - Qiuming Yao
- Department of Endocrinology, Jinshan Hospital of Fudan University, Shanghai, China
| | - Qian Li
- Department of Endocrinology, Jinshan Hospital of Fudan University, Shanghai, China
| | - Kaida Mu
- Department of Endocrinology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Jin-An Zhang
- Department of Endocrinology, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| |
Collapse
|
39
|
Silva-Vilches C, Ring S, Schrader J, Clausen BE, Probst HC, Melchior F, Schild H, Enk A, Mahnke K. Production of Extracellular Adenosine by CD73 + Dendritic Cells Is Crucial for Induction of Tolerance in Contact Hypersensitivity Reactions. J Invest Dermatol 2018; 139:541-551. [PMID: 30393085 DOI: 10.1016/j.jid.2018.10.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 10/02/2018] [Accepted: 10/15/2018] [Indexed: 02/02/2023]
Abstract
Dendritic cells (DCs) express the ecto-5'-nucleotidase CD73 that generates immunosuppressive adenosine (Ado) by dephosphorylation of extracellular Ado monophosphate and diphosphate. To investigate whether CD73-derived Ado has immune-suppressive activity, 2,4-dinitrothiocyanobenzene (DNTB) was applied to skin of wild-type (WT) or CD73-deficient (CD73-/-) mice, followed by sensitization and challenge with 2,4-dinitrofluorobenzene. In this model, we show the induction of tolerance by DNTB against 2,4-dinitrofluorobenzene only in WT but not in CD73-/- mice. Analysis of skin DCs showed increased expression of CD73 after application of DNTB in WT mice. That was accompanied by elevated concentrations of extracellular Ado in the lymph node. Moreover, T cells expressed markers for anergy, namely EGR2 and NDRG1 in DNTB-treated WT mice and they exhibited impaired proliferation upon ex vivo re-stimulation. Similarly, in vitro we observed that Ado-producing WT DCs, but not CD73-/- DCs, rendered transgenic T cells from OTII mice (OTII T cells) hyporeactive, decreased their T-cell costimulatory signaling, and induced up-regulation of EGR2 and NDRG1. Thus, these data show that expression of CD73 by DCs, which triggers elevated levels of extracellular Ado, is a crucial mechanism for the induction of anergic T cells and tolerance.
Collapse
Affiliation(s)
- Cinthia Silva-Vilches
- Department of Dermatology, Ruprecht-Karls-University Heidelberg, Heidelberg, Germany
| | - Sabine Ring
- Department of Dermatology, Ruprecht-Karls-University Heidelberg, Heidelberg, Germany
| | - Jürgen Schrader
- Institute for Molecular Cardiology, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Björn E Clausen
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Hans-Christian Probst
- Institute for Immunology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Felix Melchior
- Institute for Immunology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Hansjörg Schild
- Institute for Immunology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Alexander Enk
- Department of Dermatology, Ruprecht-Karls-University Heidelberg, Heidelberg, Germany
| | - Karsten Mahnke
- Department of Dermatology, Ruprecht-Karls-University Heidelberg, Heidelberg, Germany.
| |
Collapse
|
40
|
Zacca ER, Onofrio LI, Acosta CDV, Ferrero PV, Alonso SM, Ramello MC, Mussano E, Onetti L, Cadile II, Stancich MI, Taboada Bonfanti MC, Montes CL, Acosta Rodríguez EV, Gruppi A. PD-L1 + Regulatory B Cells Are Significantly Decreased in Rheumatoid Arthritis Patients and Increase After Successful Treatment. Front Immunol 2018; 9:2241. [PMID: 30327652 PMCID: PMC6174216 DOI: 10.3389/fimmu.2018.02241] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 09/10/2018] [Indexed: 01/18/2023] Open
Abstract
Background: B cells play an important role in the development and maintenance of rheumatoid arthritis (RA). Although IL-10-producing B cells represent a major subset of regulatory B cells (Bregs) able to suppress autoimmune and inflammatory responses, recent reports showed that B cell-mediated immune suppression may also occur independent of IL-10. For instance, B cells can modulate T cell immune responses through the expression of regulatory molecules such as PD-L1. So far, PD-L1-expressing B cells have not been analyzed in RA patients. Objective: To analyze the frequency of PD-L1-expressing B cells in the peripheral blood of RA patients compared to healthy controls (HC) matched for sex and age, their function on T cell response and their changes in response to therapy. Methods: Fresh peripheral blood B cells from RA patients and HC were characterized by flow cytometry and their functionality assessed in a co-culture system with autologous T cells. Results: The frequencies of CD19+PD-L1+ B cells, CD24hiCD38-PD-L1+ and CD24hiCD38hiPD-L1+ B cells were significantly lower in untreated RA patients than in HC. In a follow-up study, the frequencies of PD-L1+ B cells (CD19+PD-L1+ B cells, CD24hiCD38-PD-L1+ and CD24hiCD38hiPD-L1+ B cells) increased significantly after treatment in good responder patients, although the frequency of total CD24hiCD38hi B cells decreased. CD19+ B cells from untreated RA patients and HC upregulated PD-L1 expression similarly upon stimulation with CpG plus IL-2 and were able to suppress, in vitro, CD8+ T cell proliferation and cytokine production in a PD-L1-dependent manner. Conclusions: Our results show that PD-L1+ B cells exhibiting T cell suppressive capacity are significantly decreased in untreated RA patients but increase in response to successful treatment. PD-L1 expression on B cells from RA patients can be modulated in vitro and PD-L1+ B cells could thus provide new perspectives for future treatment strategies.
Collapse
Affiliation(s)
- Estefanía R Zacca
- Laboratorio de Inmunología, Hospital Nacional de Clínicas (HNC), Universidad Nacional de Córdoba (UNC), Córdoba, Argentina
| | - Luisina I Onofrio
- Laboratorio de Inmunología, Hospital Nacional de Clínicas (HNC), Universidad Nacional de Córdoba (UNC), Córdoba, Argentina
| | - Cristina D V Acosta
- Laboratorio de Inmunología, Hospital Nacional de Clínicas (HNC), Universidad Nacional de Córdoba (UNC), Córdoba, Argentina
| | - Paola V Ferrero
- Laboratorio de Inmunología, Hospital Nacional de Clínicas (HNC), Universidad Nacional de Córdoba (UNC), Córdoba, Argentina
| | - Sergio M Alonso
- Laboratorio de Inmunología, Hospital Nacional de Clínicas (HNC), Universidad Nacional de Córdoba (UNC), Córdoba, Argentina
| | - María C Ramello
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Facultad de Ciencias Químicas, UNC, Córdoba, Argentina
| | - Eduardo Mussano
- Servicio de Reumatología. Hospital Nacional de Clínicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Laura Onetti
- Servicio de Reumatología. Hospital Nacional de Clínicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Isaac I Cadile
- Servicio de Reumatología. Hospital Nacional de Clínicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Maria I Stancich
- Servicio de Reumatología. Hospital Nacional de Clínicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Maria C Taboada Bonfanti
- Servicio de Reumatología. Hospital Nacional de Clínicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Carolina L Montes
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Facultad de Ciencias Químicas, UNC, Córdoba, Argentina
| | - Eva V Acosta Rodríguez
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Facultad de Ciencias Químicas, UNC, Córdoba, Argentina
| | - Adriana Gruppi
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI-CONICET), Facultad de Ciencias Químicas, UNC, Córdoba, Argentina
| |
Collapse
|
41
|
Langdon K, Haleagrahara N. Regulatory T-cell dynamics with abatacept treatment in rheumatoid arthritis. Int Rev Immunol 2018; 37:206-214. [PMID: 29757670 DOI: 10.1080/08830185.2018.1465943] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The progressive damage in rheumatoid arthritis (RA) has been linked to an increase in inflammatory Th1/Th17 cells and a decrease in number or function of immunomodulatory regulatory T cells (Tregs). Many therapies that are effective in RA are shown to affect Th1/Th17 cells and/or Tregs. One such therapy, abatacept, utilizes a physiologic immunomodulatory molecule called cytotoxic lymphocyte antigen-4 (CTLA-4) which causes contact-dependent inhibition of inflammatory T-cell activation. Recent advances in CTLA-4 research has uncovered the method by which this occurs physiologically but the actions of the CTLA-4Ig fusion protein are still not fully understood. The reported effects of the drug on Treg population number and suppressor function have been very mixed. In this review, we will discuss the current literature surrounding the effects of abatacept in rheumatoid arthritis and explore potential explanations for the differences in results. Future opportunities in this area include contributions to a unified definition for different immune cell populations, LAG3+ Tregs which may pose an avenue for further study or the stratification of patients with regards to their specific disease characteristics, resulting in optimized treatment for disease remission.
Collapse
Affiliation(s)
- Kane Langdon
- a College of Medicine and Dentistry , James Cook University , Douglas , Townsville , Australia
| | - Nagaraja Haleagrahara
- b College of Public Health, Medicine and Veterinary Sciences , James Cook University , Douglas , Townsville , Australia
| |
Collapse
|
42
|
|
43
|
Okamura T, Yamamoto K, Fujio K. Early Growth Response Gene 2-Expressing CD4 +LAG3 + Regulatory T Cells: The Therapeutic Potential for Treating Autoimmune Diseases. Front Immunol 2018. [PMID: 29535721 PMCID: PMC5834469 DOI: 10.3389/fimmu.2018.00340] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Regulatory T cells (Tregs) are necessary for the maintenance of immune tolerance. Tregs are divided into two major populations: one is thymus derived and the other develops in the periphery. Among these Tregs, CD4+CD25+ Tregs, which mainly originate in the thymus, have been extensively studied. Transcription factor Foxp3 is well known as a master regulatory gene for the development and function of CD4+CD25+ Tregs. On the other hand, peripheral Tregs consist of distinct cell subsets including Foxp3-dependent extrathymically developed Tregs and interleukin (IL)-10-producing type I regulatory T (Tr1) cells. Lymphocyte activation gene 3 (LAG3) and CD49b are reliable cell surface markers for Tr1 cells. CD4+CD25−LAG3+ Tregs (LAG3+ Tregs) develop in the periphery and produce a large amount of IL-10. LAG3+ Tregs characteristically express the early growth response gene 2 (Egr2), a zinc-finger transcription factor, and exhibit its suppressive activity in a Foxp3-independent manner. Although Egr2 was known to be essential for hindbrain development and myelination of the peripheral nervous system, recent studies revealed that Egr2 plays vital roles in the induction of T cell anergy and also the suppressive activities of LAG3+ Tregs. Intriguingly, forced expression of Egr2 converts naive CD4+ T cells into IL-10-producing Tregs that highly express LAG3. Among the four Egr gene family members, Egr3 is thought to compensate for the function of Egr2. Recently, we reported that LAG3+ Tregs suppress humoral immune responses via transforming growth factor β3 production in an Egr2- and Egr3-dependent manner. In this review, we focus on the role of Egr2 in Tregs and also discuss its therapeutic potential for the treatment of autoimmune diseases.
Collapse
Affiliation(s)
- Tomohisa Okamura
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Max Planck-The University of Tokyo Center for Integrative Inflammology, The University of Tokyo, Tokyo, Japan
| | - Kazuhiko Yamamoto
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Max Planck-The University of Tokyo Center for Integrative Inflammology, The University of Tokyo, Tokyo, Japan.,Laboratory for Autoimmune Diseases, Center for Integrative Medical Sciences, RIKEN, Yokohama, Japan
| | - Keishi Fujio
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
44
|
Fonseka CY, Rao DA, Raychaudhuri S. Leveraging blood and tissue CD4+ T cell heterogeneity at the single cell level to identify mechanisms of disease in rheumatoid arthritis. Curr Opin Immunol 2017; 49:27-36. [PMID: 28888129 PMCID: PMC5705469 DOI: 10.1016/j.coi.2017.08.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 08/12/2017] [Indexed: 12/17/2022]
Abstract
CD4+ T cells have been long known to play an important role in the pathogenesis of rheumatoid arthritis (RA), but the specific cell populations and states that drive the disease have been challenging to identify with low dimensional single cell data and bulk assays. The advent of high dimensional single cell technologies-like single cell RNA-seq or mass cytometry-has offered promise to defining key populations, but brings new methodological and statistical challenges. Recent single cell profiling studies have revealed a broad diversity of cell types among CD4+ T cells, identifying novel populations that are expanded or altered in RA. Here, we will review recent findings on CD4+ T cell heterogeneity and RA that have come from single cell profiling studies and discuss the best practices for conducting these studies.
Collapse
Affiliation(s)
- Chamith Y Fonseka
- Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA; Division of Genetics, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA; Program in Medical and Population Genetics, Broad Institute of Massachusetts Technical Institute and Harvard University, Cambridge, MA 02138, USA; Center for Data Sciences, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| | - Deepak A Rao
- Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Soumya Raychaudhuri
- Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA; Division of Genetics, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA; Program in Medical and Population Genetics, Broad Institute of Massachusetts Technical Institute and Harvard University, Cambridge, MA 02138, USA; Center for Data Sciences, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA; Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| |
Collapse
|
45
|
McCarthy MW, Walsh TJ. Checkpoint inhibitors and the risk of infection. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2017. [DOI: 10.1080/23808993.2017.1380517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Matthew William McCarthy
- Hospital Medicine, Joan and Sanford I Weill Medical College of Cornell University, New York, NY, USA
| | - Thomas J. Walsh
- Transplantation-Oncology Infectious Diseases Program, Weill Cornell Medical Center, New York, NY, USA
| |
Collapse
|
46
|
Sergi C, Shen F, Lim DW, Liu W, Zhang M, Chiu B, Anand V, Sun Z. Cardiovascular dysfunction in sepsis at the dawn of emerging mediators. Biomed Pharmacother 2017; 95:153-160. [PMID: 28841455 DOI: 10.1016/j.biopha.2017.08.066] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 08/03/2017] [Accepted: 08/13/2017] [Indexed: 12/14/2022] Open
Abstract
Subcellular dysfunction and impaired metabolism derived from the complex interaction of cytokines and mediators with cellular involvement are on the basis of the cardiovascular response to sepsis. The lethal consequences of an infection are intimately related to its ability to spread to other organ sites and the immune system of the host. About one century ago, William Osler (1849-1919), a Canadian physician, remarkably defined the sequelae of the host response in sepsis: "except on few occasions, the patient appears to die from the body's response to infection rather than from it." Cardiac dysfunction has received considerable attention to explain the heart failure in patients progressing from infection to sepsis, but our understanding of the processes remains limited. In fact, most concepts are linked to a mechanical concept of the sarcomeric structure, and physiological data seems to be often disconnected. Cytokines, prostanoids, and nitric oxide release are high direct impact factors, but coronary circulation and cardiomyocyte physiology also play a prominent role in modulating the effects of monocyte adhesion and infiltration. Damage-associated molecular patterns (DAMPs) and pathogen-associated molecular patterns (PAMPs) are involved in the host response. The identification of microRNAs, as well as the cyclic activation of the inflammatory cascade, has further added complexity to the scene. In this review, we delineate the current concepts of cellular dysfunction of the cardiomyocyte in the setting of sepsis and consider potential therapeutic strategies.
Collapse
Affiliation(s)
- Consolato Sergi
- Institute of Biomedical and Pharmaceutical Sciences, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, 430068, PR China; Department of Orthopedics, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, Hubei, PR China; Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada; Stollery Children's Hospital, University Alberta Hospital, Edmonton, AB, Canada.
| | - Fan Shen
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - David W Lim
- Department of Surgery, University of Alberta, Edmonton, AB, Canada
| | - Weiyong Liu
- Department of Clinical Laboratory, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Mingyong Zhang
- Department of Orthopedics, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, Hubei, PR China
| | - Brian Chiu
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - Vijay Anand
- Department of Critical Care Medicine, University of Alberta, Edmonton, AB, Canada
| | - Ziyong Sun
- Department of Clinical Laboratory, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| |
Collapse
|