1
|
Wang X, Ni T, Miao J, Huang X, Feng Z. The role and mechanism of triptolide, a potential new DMARD, in the treatment of rheumatoid arthritis. Ageing Res Rev 2025; 104:102643. [PMID: 39722411 DOI: 10.1016/j.arr.2024.102643] [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: 08/31/2024] [Revised: 12/09/2024] [Accepted: 12/16/2024] [Indexed: 12/28/2024]
Abstract
Triptolide (TP) is the primary pharmacological component of Tripterygium Glycosides (TG), which has anti-inflammatory, antiproliferative, and immunosuppressive properties, among other pharmacological actions, and has excellent potential for developing into a new DMARD. We have reviewed the effects and mechanisms of TP on immunosuppression, inhibiting synovial proliferation, and preventing articular bone destruction in the treatment of rheumatoid arthritis (RA), which is a common disease in the elderly in this paper. We have found that TP has regulatory effects on multiple vital cells in the above-mentioned pathological process of RA, such as monocytes/macrophages, dendritic cells, T cells, fibroblast-like synoviocytes, and osteoclasts. We also found that TP can regulate multiple key signaling pathways such as NF-κB, JAK/STAT, and MAPK through various molecular regulatory mechanisms, achieving regulatory effects on numerous phenotypes of the above-mentioned vital cells.
Collapse
Affiliation(s)
- Xiwen Wang
- The First Clinical Medical College, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Nanjing 210023, China
| | - Tianyang Ni
- The First Clinical Medical College, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Nanjing 210023, China
| | - Jianru Miao
- The First Clinical Medical College, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Nanjing 210023, China
| | - Xinyao Huang
- The First Clinical Medical College, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Nanjing 210023, China
| | - Zhe Feng
- The First Clinical Medical College, Nanjing University of Chinese Medicine, No. 138 Xianlin Avenue, Nanjing 210023, China.
| |
Collapse
|
2
|
Li N, Xu T, Wu Z, Zhao Y, Ruan M, Xu H, Chen W, Wang H, Wang S, Wang Y, Liang Q. Arabinogalactan from Cynanchum atratum induces tolerogenic dendritic cells in gut to restrain autoimmune response and alleviate collagen-induced arthritis in mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156269. [PMID: 39586124 DOI: 10.1016/j.phymed.2024.156269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 11/08/2024] [Accepted: 11/15/2024] [Indexed: 11/27/2024]
Abstract
BACKGROUND Rheumatoid arthritis (RA) is an autoimmune disease characterized by multiple joints lesions. Tolerogenic dendritic cells (tolDCs) play crucial roles in maintaining immune homeostasis. The immunomodulatory activity of plant-derived arabinogalactan (AGs) has been well investigated, however, whether AGs could suppress autoimmune responses by inducing tolDCs is remain unclear. DESIGN Collagen-induced arthritis (CIA, a mouse model of RA) mice were utilized to ascertain the role of AGs (obtained from Cynanchum atratum) in autoimmune responses. An antibiotic cocktail was administered to eliminate gut microbiota. Germ-free (GF) and Toll-like receptor 2 (TLR2) knockout mice were used to determine the function of AGs in intestinal immune cells. RESULTS The oral administration of dietary AGs substantially reduced the severity of CIA and rebalanced the ratio of regulatory T cells (Tregs) to T helper 17 (Th17) cells. Although the antibiotic cocktail depleted the mice's gut microbiota, AGs had a therapeutic effect on their CIA. AGs restored Treg/Th17 homeostasis by inducing CD103+ tolDCs, regardless of the gut microbiota of the GF mice. Coculture experiments confirmed that AGs induced tolDCs and transforming growth factor β (TGF-β) secretion, leading to Treg amplification. RNA sequencing and TLR2 knockout experiments revealed that AGs induced tolDCs through a TLR2-mediated mechanism. Preventive interventions with AGs established a tolerogenic intestinal immune microenvironment, which delayed the onset and progression of CIA. AGs functioned synergistically with tofacitinib, a JAK inhibitor, to effectively restore Treg/Th17 balance and alleviate CIA. CONCLUSION This study introduces a novel microbiota-independent mechanism through which soluble dietary AGs inhibit systemic autoimmune responses. Our findings provide insights into the supplementation of dietary AGs in patients with preclinical or progressive RA.
Collapse
Affiliation(s)
- Ning Li
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, PR China; Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, PR China
| | - Tianhao Xu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, PR China; Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, PR China
| | - Zhaoshun Wu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, PR China; Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, PR China
| | - Yuchen Zhao
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, PR China; Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, PR China
| | - Ming Ruan
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, PR China; Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, PR China
| | - Hao Xu
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, PR China; Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, PR China
| | - Weihao Chen
- The MOE Key Laboratory for Standardization of Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, PR China
| | - Huijun Wang
- The MOE Key Laboratory for Standardization of Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, PR China
| | - Shunchun Wang
- The MOE Key Laboratory for Standardization of Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, PR China.
| | - Yongjun Wang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, PR China; Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, PR China.
| | - Qianqian Liang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, PR China; Spine Institute, Shanghai University of Traditional Chinese Medicine, 725 Wan-Ping South Road, Shanghai 200032, PR China.
| |
Collapse
|
3
|
Arve-Butler S, Moorman CD. A comprehensive overview of tolerogenic vaccine adjuvants and their modes of action. Front Immunol 2024; 15:1494499. [PMID: 39759532 PMCID: PMC11695319 DOI: 10.3389/fimmu.2024.1494499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 11/29/2024] [Indexed: 01/07/2025] Open
Abstract
Tolerogenic vaccines represent a therapeutic approach to induce antigen-specific immune tolerance to disease-relevant antigens. As general immunosuppression comes with significant side effects, including heightened risk of infections and reduced anti-tumor immunity, antigen-specific tolerance by vaccination would be game changing in the treatment of immunological conditions such as autoimmunity, anti-drug antibody responses, transplantation rejection, and hypersensitivity. Tolerogenic vaccines induce antigen-specific tolerance by promoting tolerogenic antigen presenting cells, regulatory T cells, and regulatory B cells, or by suppressing or depleting antigen-specific pathogenic T and B cells. The design of tolerogenic vaccines vary greatly, but they all deliver a disease-relevant antigen with or without a tolerogenic adjuvant. Tolerogenic adjuvants are molecules which mediate anti-inflammatory or immunoregulatory effects and enhance vaccine efficacy by modulating the immune environment to favor a tolerogenic immune response to the vaccine antigen. Tolerogenic adjuvants act through several mechanisms, including immunosuppression, modulation of cytokine signaling, vitamin signaling, and modulation of immunological synapse signaling. This review seeks to provide a comprehensive examination of tolerogenic adjuvants currently utilized in tolerogenic vaccines, describing their mechanism of action and examples of their use in human clinical trials and animal models of disease.
Collapse
Affiliation(s)
- Sabine Arve-Butler
- Amgen R&D Postdoctoral Fellows Program, Amgen Inc, South San Francisco, CA, United States
- Amgen Research, Amgen Inc., South San Francisco, CA, United States
| | | |
Collapse
|
4
|
Beckers D, Jainarayanan AK, Dustin ML, Capera J. T Cell Resistance: On the Mechanisms of T Cell Non-activation. Immune Netw 2024; 24:e42. [PMID: 39801736 PMCID: PMC11711127 DOI: 10.4110/in.2024.24.e42] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 10/28/2024] [Accepted: 11/12/2024] [Indexed: 01/16/2025] Open
Abstract
Immunological tolerance is a fundamental arm of any functioning immune system. Not only does tolerance mitigate collateral damage from host immune responses, but in doing so permits a robust response sufficient to clear infection as necessary. Yet, despite occupying such a cornerstone, research aiming to unravel the intricacies of tolerance induction is mired by interchangeable and often misused terminologies, with markers and mechanistic pathways that beg the question of redundancy. In this review we aim to define these boarders by providing new perspectives to long-standing theories of tolerance. Given the central role of T cells in enforcing immune cascades, in this review we choose to explore immunological tolerance through the perspective of T cell 'resistance to activation,' to delineate the contexts in which one tolerance mechanism has evolved over the other. By clarifying the important biological markers and cellular players underpinning T cell resistance to activation, we aim to encourage more purposeful and directed research into tolerance and, more-over, potential therapeutic strategies in autoimmune diseases and cancer. The tolerance field is in much need of reclassification and consideration, and in this review, we hope to open that conversation.
Collapse
Affiliation(s)
- Daniel Beckers
- The Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics Rheumatology & Musculoskeletal Sciences, University of Oxford, Oxford OX3 7FY, United Kingdom
| | - Ashwin K. Jainarayanan
- The Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics Rheumatology & Musculoskeletal Sciences, University of Oxford, Oxford OX3 7FY, United Kingdom
| | - Michael L. Dustin
- The Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics Rheumatology & Musculoskeletal Sciences, University of Oxford, Oxford OX3 7FY, United Kingdom
| | - Jesusa Capera
- The Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics Rheumatology & Musculoskeletal Sciences, University of Oxford, Oxford OX3 7FY, United Kingdom
| |
Collapse
|
5
|
Song Y, Li J, Wu Y. Evolving understanding of autoimmune mechanisms and new therapeutic strategies of autoimmune disorders. Signal Transduct Target Ther 2024; 9:263. [PMID: 39362875 PMCID: PMC11452214 DOI: 10.1038/s41392-024-01952-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 07/09/2024] [Accepted: 08/07/2024] [Indexed: 10/05/2024] Open
Abstract
Autoimmune disorders are characterized by aberrant T cell and B cell reactivity to the body's own components, resulting in tissue destruction and organ dysfunction. Autoimmune diseases affect a wide range of people in many parts of the world and have become one of the major concerns in public health. In recent years, there have been substantial progress in our understanding of the epidemiology, risk factors, pathogenesis and mechanisms of autoimmune diseases. Current approved therapeutic interventions for autoimmune diseases are mainly non-specific immunomodulators and may cause broad immunosuppression that leads to serious adverse effects. To overcome the limitations of immunosuppressive drugs in treating autoimmune diseases, precise and target-specific strategies are urgently needed. To date, significant advances have been made in our understanding of the mechanisms of immune tolerance, offering a new avenue for developing antigen-specific immunotherapies for autoimmune diseases. These antigen-specific approaches have shown great potential in various preclinical animal models and recently been evaluated in clinical trials. This review describes the common epidemiology, clinical manifestation and mechanisms of autoimmune diseases, with a focus on typical autoimmune diseases including multiple sclerosis, type 1 diabetes, rheumatoid arthritis, systemic lupus erythematosus, and sjögren's syndrome. We discuss the current therapeutics developed in this field, highlight the recent advances in the use of nanomaterials and mRNA vaccine techniques to induce antigen-specific immune tolerance.
Collapse
Affiliation(s)
- Yi Song
- Institute of Immunology, PLA, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jian Li
- Chongqing International Institute for Immunology, Chongqing, China.
| | - Yuzhang Wu
- Institute of Immunology, PLA, Third Military Medical University (Army Medical University), Chongqing, China.
- Chongqing International Institute for Immunology, Chongqing, China.
| |
Collapse
|
6
|
Stoppelenburg AJ, Schreibelt G, Koeneman B, Welsing P, Breman EJ, Lammers L, de Goede A, Duiveman-de Boer T, van Eden W, Leufkens P, de Vries IJM, Broere F, van Laar JM. Design of TOLERANT: phase I/II safety assessment of intranodal administration of HSP70/mB29a self-peptide antigen-loaded autologous tolerogenic dendritic cells in patients with rheumatoid arthritis. BMJ Open 2024; 14:e078231. [PMID: 39266308 PMCID: PMC11409275 DOI: 10.1136/bmjopen-2023-078231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 08/21/2024] [Indexed: 09/14/2024] Open
Abstract
INTRODUCTION In rheumatoid arthritis (RA), immunosuppressive therapies may achieve symptomatic relief, but do not induce long-term, drug-free remission. Meanwhile, the lifelong use of immunosuppressive drugs confers increased risk for malignancy and infections. As such, there is an unmet need for novel treatments that selectively target the pathogenic immune response in RA by inducing tolerance to autoantigens. Autologous cell therapy using antigen-loaded tolerogenic dendritic cells (tolDCs) aims to reinstate autoantigen-specific immunological tolerance in RA and could potentially meet this need. METHODS AND ANALYSIS We report here the design of the phase I/II, investigator-initiated, open-label, dose-escalation trial TOLERANT. In this study, we will evaluate the intranodal administration of tolDCs in patients with RA that are in remission under immunosuppressive therapy. The tolDCs in this trial are loaded with the heat shock protein 70-derived peptide mB29a, which is an effective surrogate autoantigen in animal models of arthritis. Within this study, three dose-escalation cohorts (two intranodal injections of 5×106, 10×106 and 15×106 tolDCs), each consisting of three patients, are evaluated to identify the highest safe dose (recommended dose), and an extension cohort of nine patients will be treated with the recommended dose. The (co-)primary endpoints of this study are safety and feasibility, which we assess by the number of AEs and the successful production of tolDCs. The secondary endpoints include the immunological effects of the treatment, which we assess with a variety of high-dimensional and antigen-specific immunological assays. Clinical effects are exploratory outcomes. ETHICS AND DISSEMINATION Ethical approval for this study has been obtained from the Netherlands Central Committee on Research Involving Human Subjects. The outcomes of the trial will be disseminated through publications in open-access, peer-reviewed scientific journals, scientific conferences and to patient associations. TRIAL REGISTRATION NUMBERS NCT05251870; 2019-003620-20 (EudraCT); NL71296.000.20 (CCMO register).
Collapse
Affiliation(s)
- Arie Jan Stoppelenburg
- Department of Rheumatology & Clinical Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
- Department of Biomolecular Health Sciences, Utrecht University Faculty of Veterinary Medicine, Utrecht, The Netherlands
| | - Gerty Schreibelt
- Department of Medical BioSciences, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Bouke Koeneman
- Department of Medical BioSciences, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Paco Welsing
- Department of Rheumatology & Clinical Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Evert-Jan Breman
- Department of Clinical Pharmacy, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Laureen Lammers
- Department of Clinical Pharmacy, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Anna de Goede
- Department of Pharmacy, Radboud University Medical Centre, Nijmegen, The Netherlands
| | | | - Willem van Eden
- Department of Biomolecular Health Sciences, Utrecht University Faculty of Veterinary Medicine, Utrecht, The Netherlands
- Trajectum Pharma B.V, Utrecht, The Netherlands
| | | | - I Jolanda M de Vries
- Department of Medical BioSciences, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Femke Broere
- Department of Biomolecular Health Sciences, Utrecht University Faculty of Veterinary Medicine, Utrecht, The Netherlands
| | - Jacob M van Laar
- Department of Rheumatology & Clinical Immunology, University Medical Centre Utrecht, Utrecht, The Netherlands
| |
Collapse
|
7
|
Wang S, Xu Y, Wang L, Lin J, Xu C, Zhao X, Zhang H. TolDC Restores the Balance of Th17/Treg via Aryl Hydrocarbon Receptor to Attenuate Colitis. Inflamm Bowel Dis 2024; 30:1546-1555. [PMID: 38431309 DOI: 10.1093/ibd/izae022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Indexed: 03/05/2024]
Abstract
BACKGROUND Tolerogenic dendritic cells (TolDCs) have been evidenced to trigger regulatory T cell's (Treg's) differentiation and be involved in the pathogenesis of Crohn's disease (CD). Aryl hydrocarbon receptor (AhR) plays a crucial role in the differentiation of TolDCs, although the mechanism remains vague. This study aimed to evaluate the role of AhR in TolDCs formation, which may affect Th17/Treg balance in CD. METHODS Colon biopsy specimens were obtained from healthy controls and patients with CD. Wild type (WT) and AhR-/- mice were induced colitis by drinking dextran sulphate sodium (DSS) with or without 6-formylindolo 3,2-b carbazole (FICZ) treatment. Wild type and AhR-/- bone marrow-derived cells (BMDCs) were cultured under TolDCs polarization condition. Ratios of DCs surface markers were determined by flow cytometry. Enzyme-linked immunosorbent assay (ELISA) was performed to quantify the levels of interleukin (IL)-1β, transforming growth factor (TGF)-β and IL-10. Tolerogenic dendritic cells differentiated from BMDCs of WT or AhR-/- mice were adoptively transferred to DSS-induced WT colitis mice. RESULTS Patients with CD showed less AhR expression and activation in their inflamed colon regions. Compared with WT mice, AhR-/- mice experienced more severe colitis. Tolerogenic dendritic cells and Tregs were both decreased in the colon of AhR-/- colitis mice, while Th17 cells were upregulated. In vitro, compared with WT DCs, AhR-deficient DCs led to less TolDC formation. Furthermore, intestinal inflammation in WT colitis mice, which transferred with AhR-/- TolDCs, showed no obvious improvement compared with those transferred with WT TolDCs, as evidenced by no rescues of Th17/Treg balance. CONCLUSIONS Activation of AhR attenuates experimental colitis by modulating the balance of TolDCs and Th17/Treg. The AhR modulation of TolDCs may be a viable therapeutic approach for CD.
Collapse
Affiliation(s)
- Shu Wang
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu Province, People's Republic of China
| | - Ying Xu
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu Province, People's Republic of China
| | - Lu Wang
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu Province, People's Republic of China
| | - Junjie Lin
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu Province, People's Republic of China
| | - Chenjing Xu
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu Province, People's Republic of China
| | - Xiaojing Zhao
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu Province, People's Republic of China
| | - Hongjie Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu Province, People's Republic of China
| |
Collapse
|
8
|
Benne N, Ter Braake D, Porenta D, Lau CYJ, Mastrobattista E, Broere F. Autoantigen-Dexamethasone Conjugate-Loaded Liposomes Halt Arthritis Development in Mice. Adv Healthc Mater 2024; 13:e2304238. [PMID: 38295848 DOI: 10.1002/adhm.202304238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/23/2024] [Indexed: 02/13/2024]
Abstract
There is no curative treatment for chronic auto-inflammatory diseases including rheumatoid arthritis, and current treatments can induce off-target side effects due to systemic immune suppression. This work has previously shown that dexamethasone-pulsed tolerogenic dendritic cells loaded with the arthritis-specific antigen human proteoglycan can suppress arthritis development in a proteoglycan-induced arthritis mouse model. To circumvent ex vivo dendritic cell culture, and enhance antigen-specific effects, drug delivery vehicles, such as liposomes, provide an interesting approach. Here, this work uses anionic 1,2-distearoyl-sn-glycero-3-phosphoglycerol liposomes with enhanced loading of human proteoglycan-dexamethasone conjugates by cationic lysine tetramer addition. Antigen-pulsed tolerogenic dendritic cells induced by liposomal dexamethasone in vitro enhanced antigen-specific regulatory T cells to a similar extent as dexamethasone-induced tolerogenic dendritic cells. In an inflammatory adoptive transfer model, mice injected with antigen-dexamethasone liposomes have significantly higher antigen-specific type 1 regulatory T cells than mice injected with antigen only. The liposomes significantly inhibit the progression of arthritis compared to controls in preventative and therapeutic proteoglycan-induced arthritis mouse models. This coincides with systemic tolerance induction and an increase in IL10 expression in the paws of mice. In conclusion, a single administration of autoantigen and dexamethasone-loaded liposomes seems to be a promising antigen-specific treatment strategy for arthritis in mice.
Collapse
Affiliation(s)
- Naomi Benne
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584 CL, The Netherlands
| | - Daniëlle Ter Braake
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584 CL, The Netherlands
| | - Deja Porenta
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584 CL, The Netherlands
- Utrecht Institute for Pharmaceutical Sciences, Department of Pharmaceutics, Faculty of Science, Utrecht University, Utrecht, 3584 CG, The Netherlands
| | - Chun Yin Jerry Lau
- Utrecht Institute for Pharmaceutical Sciences, Department of Pharmaceutics, Faculty of Science, Utrecht University, Utrecht, 3584 CG, The Netherlands
- Department of Materials Engineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Enrico Mastrobattista
- Utrecht Institute for Pharmaceutical Sciences, Department of Pharmaceutics, Faculty of Science, Utrecht University, Utrecht, 3584 CG, The Netherlands
| | - Femke Broere
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584 CL, The Netherlands
| |
Collapse
|
9
|
Jia C, Wang Y, Wang Y, Cheng M, Dong W, Wei W, Zhao Y, Chang Y. TDO2-overexpressed Dendritic Cells Possess Tolerogenicity and Ameliorate Collagen-induced Arthritis by Modulating the Th17/Regulatory T Cell Balance. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:941-950. [PMID: 38294261 DOI: 10.4049/jimmunol.2300442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 01/09/2024] [Indexed: 02/01/2024]
Abstract
Tolerogenic dendritic cells are promising for restoring immune homeostasis and may be an alternative therapy for autoimmune diseases such as rheumatoid arthritis. The kynurenine pathway is a vital mechanism that induces tolerance in dendritic cells (DCs). Tryptophan 2,3-dioxygenase (TDO2) is an important rate-limiting enzyme in the kynurenine pathway and participates in immune regulation. However, the role of TDO2 in shaping the tolerogenic phenotypes of DCs remains unclear. In this study, we investigated the effects and mechanisms of TDO2-overexpressed DCs in regulating the T cell balance both in vivo and in vitro. TDO2-overexpressed DC2.4 and TDO2-/- mouse bone marrow-derived DCs (BMDCs) were generated to verify the role of TDO2 in DC maturation and functionality. TDO2 overexpression in BMDCs via PGE2 treatment exhibited an immature phenotype and tolerogenic state, whereas TDO2-/- BMDCs exhibited a mature phenotype and a proinflammatory state. Furthermore, transplant of TDO2-overexpressed BMDCs alleviated collagen-induced arthritis severity in mice, which was correlated with a reduction in Th17 populations and an increase in regulatory T cells. Collectively, these results indicate that TDO2 plays an important role in the tolerogenic phenotype and may be a promising target for the generation tolerogenic DCs for rheumatoid arthritis treatment.
Collapse
Affiliation(s)
- Chengyan Jia
- Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, China
| | - Yueye Wang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, China
| | - Yi Wang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, China
| | - Meng Cheng
- Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, China
| | - Weibo Dong
- Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, China
| | - Wei Wei
- Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, China
| | - Yingjie Zhao
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yan Chang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, China
| |
Collapse
|
10
|
Mehta JM, Hiremath SC, Chilimba C, Ghasemi A, Weaver JD. Translation of cell therapies to treat autoimmune disorders. Adv Drug Deliv Rev 2024; 205:115161. [PMID: 38142739 PMCID: PMC10843859 DOI: 10.1016/j.addr.2023.115161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/05/2023] [Accepted: 12/15/2023] [Indexed: 12/26/2023]
Abstract
Autoimmune diseases are a diverse and complex set of chronic disorders with a substantial impact on patient quality of life and a significant global healthcare burden. Current approaches to autoimmune disease treatment comprise broadly acting immunosuppressive drugs that lack disease specificity, possess limited efficacy, and confer undesirable side effects. Additionally, there are limited treatments available to restore organs and tissues damaged during the course of autoimmune disease progression. Cell therapies are an emergent area of therapeutics with the potential to address both autoimmune disease immune dysfunction as well as autoimmune disease-damaged tissue and organ systems. In this review, we discuss the pathogenesis of common autoimmune disorders and the state-of-the-art in cell therapy approaches to (1) regenerate or replace autoimmune disease-damaged tissue and (2) eliminate pathological immune responses in autoimmunity. Finally, we discuss critical considerations for the translation of cell products to the clinic.
Collapse
Affiliation(s)
- Jinal M Mehta
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Shivani C Hiremath
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Chishiba Chilimba
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Azin Ghasemi
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Jessica D Weaver
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA.
| |
Collapse
|
11
|
Scotland BL, Shaw JR, Dharmaraj S, Caprio N, Cottingham AL, Joy Martín Lasola J, Sung JJ, Pearson RM. Cell and biomaterial delivery strategies to induce immune tolerance. Adv Drug Deliv Rev 2023; 203:115141. [PMID: 37980950 PMCID: PMC10842132 DOI: 10.1016/j.addr.2023.115141] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/09/2023] [Accepted: 11/10/2023] [Indexed: 11/21/2023]
Abstract
The prevalence of immune-mediated disorders, including autoimmune conditions and allergies, is steadily increasing. However, current therapeutic approaches are often non-specific and do not address the underlying pathogenic condition, often resulting in impaired immunity and a state of generalized immunosuppression. The emergence of technologies capable of selectively inhibiting aberrant immune activation in a targeted, antigen (Ag)-specific manner by exploiting the body's intrinsic tolerance pathways, all without inducing adverse side effects, holds significant promise to enhance patient outcomes. In this review, we will describe the body's natural mechanisms of central and peripheral tolerance as well as innovative delivery strategies using cells and biomaterials targeting innate and adaptive immune cells to promote Ag-specific immune tolerance. Additionally, we will discuss the challenges and future opportunities that warrant consideration as we navigate the path toward clinical implementation of tolerogenic strategies to treat immune-mediated diseases.
Collapse
Affiliation(s)
- Brianna L Scotland
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N. Pine Street, Baltimore, MD 21201, United States
| | - Jacob R Shaw
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W. Baltimore Street, Baltimore, MD 21201, United States
| | - Shruti Dharmaraj
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N. Pine Street, Baltimore, MD 21201, United States
| | - Nicholas Caprio
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N. Pine Street, Baltimore, MD 21201, United States
| | - Andrea L Cottingham
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N. Pine Street, Baltimore, MD 21201, United States
| | - Jackline Joy Martín Lasola
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W. Baltimore Street, Baltimore, MD 21201, United States
| | - Junsik J Sung
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N. Pine Street, Baltimore, MD 21201, United States
| | - Ryan M Pearson
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 N. Pine Street, Baltimore, MD 21201, United States; Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 W. Baltimore Street, Baltimore, MD 21201, United States; Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, 22 S. Greene Street, Baltimore, MD 21201, United States.
| |
Collapse
|
12
|
Moghaddam MZ, Mousavi MJ, Ghotloo S. Cell-based therapies for the treatment of rheumatoid arthritis. Immun Inflamm Dis 2023; 11:e1091. [PMID: 38018576 PMCID: PMC10664399 DOI: 10.1002/iid3.1091] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 11/01/2023] [Accepted: 11/03/2023] [Indexed: 11/30/2023] Open
Abstract
Autoimmune diseases, including rheumatoid arthritis that is the most prevalent rheumatic autoimmune disorder, affect autologous connective tissues caused by the breakdown of the self-tolerance mechanisms of the immune system. During the last two decades, cell-based therapy, including stem cells and none-stem cells has been increasingly considered as a therapeutic option in various diseases. This is partly due to the unique properties of stem cells that divide and differentiate from the specialized cells in the damaged tissue. Moreover, stem cells and none-stem cells, impose immunomodulatory properties affecting the diseases caused by immunological abnormalities such as rheumatic autoimmune disorders. In the present review, the efficacy of cell-based therapy with four main types of stem cells, including mesenchymal stem cells, hematopoietic stem cells, embryonic stem cells, and human amniotic membrane cells, as well as none-stem cells, including regulatory T cells, chimeric antigen receptor T cells, and tolerogenic dendritic cells will be evaluated. Moreover, other related issues, including safety, changes in immunological parameters, suitable choice of stem cell and none-stem cell origin, conditioning regimen, limitations, and complications will be discussed.
Collapse
Affiliation(s)
| | - Mohammad Javad Mousavi
- Department of HematologyFaculty of Allied Medicine, Bushehr University of Medical SciencesBushehrIran
| | - Somayeh Ghotloo
- Autoimmune Diseases Research CenterKashan University of Medical SciencesKashanIran
- Department of Clinical Laboratory SciencesKashan University of Medical SciencesKashanIran
| |
Collapse
|
13
|
Jonny, Putranto TA, Purnama Y, Djatmiko R, Yana ML, Sitepu EC, Irfon R. Significant improvement of systemic lupus erythematosus manifestation in children after autologous dendritic cell transfer: a case report and review of literature. Ther Adv Vaccines Immunother 2023; 11:25151355231186005. [PMID: 37719802 PMCID: PMC10501061 DOI: 10.1177/25151355231186005] [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: 12/01/2022] [Accepted: 06/12/2023] [Indexed: 09/19/2023] Open
Abstract
Dendritic cells (DC) are postulated to play a role in autoimmune diseases such as Systemic Lupus Erythematosus (SLE). We reported a 13-year-old female SLE patient who presents with chronic arthritis accompanied by persistent fever, dyspnea, sleep disturbance, headache, stomatitis, rash, and muscle weakness. The supporting examinations showed abnormal blood cell counts, positive antinuclear antibody profile, serositis, and neuropathy. Immunosuppressants failed to improve the condition. DC-based vaccine derived from autologous peripheral blood which was introduced with SARS-CoV-2 protein was given to this patient. There was a significant improvement in clinical and laboratory findings. Thus, DC immunotherapy appears to be a potential novel therapy for SLE that needs to be studied.
Collapse
Affiliation(s)
- Jonny
- Faculty of Medicine, Jakarta Veterans National Development University, Jakarta, Indonesia
- Cellcure Center, Gatot Soebroto Central Army Hospital, Jl. Abdul Rahman Saleh Raya No. 24 RT10/RW5, Jakarta 10410, Indonesia
| | | | - Yenny Purnama
- Pediatric Department Gatot Soebroto Central Army Hospital, Jakarta, Indonesia
| | - Roedi Djatmiko
- Cellcure Center, Gatot Soebroto Central Army Hospital, Jakarta, Indonesia
| | - Martina Lily Yana
- Cellcure Center, Gatot Soebroto Central Army Hospital, Jakarta, Indonesia
| | | | - Raoulian Irfon
- Cellcure Center, Gatot Soebroto Central Army Hospital, Jakarta, Indonesia
| |
Collapse
|
14
|
Suwa Y, Nagafuchi Y, Yamada S, Fujio K. The role of dendritic cells and their immunometabolism in rheumatoid arthritis. Front Immunol 2023; 14:1161148. [PMID: 37251399 PMCID: PMC10213288 DOI: 10.3389/fimmu.2023.1161148] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 04/28/2023] [Indexed: 05/31/2023] Open
Abstract
Dendritic cells (DCs) play crucial roles in the pathogenesis of rheumatoid arthritis (RA), a prototypic autoimmune disease characterized by chronic synovitis and joint destruction. Conventional dendritic cells (cDCs) with professional antigen-presenting functions are enriched in the RA synovium. In the synovium, the cDCs are activated and show both enhanced migratory capacities and T cell activation in comparison with peripheral blood cDCs. Plasmacytoid dendritic cells, another subtype of DCs capable of type I interferon production, are likely to be tolerogenic in RA. Monocyte-derived dendritic cells (moDCs), once called "inflammatory DCs", are localized in the RA synovium, and they induce T-helper 17 cell expansion and enhanced proinflammatory cytokine production. Recent studies revealed that synovial proinflammatory hypoxic environments are linked to metabolic reprogramming. Activation of cDCs in the RA synovium is accompanied by enhanced glycolysis and anabolism. In sharp contrast, promoting catabolism can induce tolerogenic DCs from monocytes. Herein, we review recent studies that address the roles of DCs and their immunometabolic features in RA. Immunometabolism of DCs could be a potential therapeutic target in RA.
Collapse
Affiliation(s)
- Yuichi Suwa
- 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
- Department of Functional Genomics and Immunological Diseases, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Saeko Yamada
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Keishi Fujio
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
15
|
Samarpita S, Rasool M. Majoon chobchini reinstates PDL-1 expression and blocks dendritic cell -T helper 17 pathogenic axis in rheumatoid arthritis animal model. Cytokine 2023; 163:156136. [PMID: 36716676 DOI: 10.1016/j.cyto.2023.156136] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 12/08/2022] [Accepted: 01/16/2023] [Indexed: 01/29/2023]
Abstract
Dendritic cells (DCs) are the critical players in the puzzle of rheumatoid arthritis (RA) disease pathogenesis. Blockade of DC activation has been shown to curtail Th17 cell differentiation and its aberrant function in RA. Recent studies have pointed to the role of the PI3K/AKT signaling axis in the maturation and activation of DCs. However, it is yet to be established how PI3K/AKT inhibition would lead to the abolishment of DC activation and Th17 cell plasticity in RA. Herein, our study decoded whether and how majoon chobchini, an unani compound, abated dendritic cell maturation and regulated the Th17/Treg paradigm in RA. Given our results, majoon chobchini conspicuously restrained MHC II, CD86 expression and, subsequently elevated PDL-1 levels in DCs in-vivo. Of note, inhibition of DC maturation by majoon chobchini, in turn, favoured suppression of the Th17 cell population while driving Treg cell development in adjuvant induced arthritic (AA) rats. Concurrently, majoon chobchini decreased the catabolic effects of IL-17 (Th17 associated cytokine) via a reciprocal increase in IL-10 (Treg associated cytokine) levels in AA rats. Mechanistically, majoon chobchini sustained FoxO1 nuclear localization signaled through dampened PI3K/AKT phosphorylation in-vitro. In concert, PDL-1 expression was heightened in majoon chobchini treated activated DCs that provides a framework for ablation of the DC-Th17 cell pathogenic axis in RA. Notwithstanding, the PI3K inhibitor LY294002 exhibited similar inhibitory effects. In essence, majoon chobchini enhanced PDL-1 expression that abolished DC maturation via regulation of the PI3K/AKT/FoxO1 axis, thereby hindering Th17 differentiation in an animal model of RA. This further warrants a clinical investigation that could validate majoon chobchini as a prospective therapeutic drug in the treatment of RA.
Collapse
Affiliation(s)
- Snigdha Samarpita
- Immunopathology Lab, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632 014, Tamil Nadu, India
| | - Mahaboobkhan Rasool
- Immunopathology Lab, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632 014, Tamil Nadu, India.
| |
Collapse
|
16
|
Van Delen M, Janssens I, Dams A, Roosens L, Ogunjimi B, Berneman ZN, Derdelinckx J, Cools N. Tolerogenic Dendritic Cells Induce Apoptosis-Independent T Cell Hyporesponsiveness of SARS-CoV-2-Specific T Cells in an Antigen-Specific Manner. Int J Mol Sci 2022; 23:15201. [PMID: 36499533 PMCID: PMC9740551 DOI: 10.3390/ijms232315201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022] Open
Abstract
Although the global pandemic caused by the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is still ongoing, there are currently no specific and highly efficient drugs for COVID-19 available, particularly in severe cases. Recent findings demonstrate that severe COVID-19 disease that requires hospitalization is associated with the hyperactivation of CD4+ and CD8+ T cell subsets. In this study, we aimed to counteract this high inflammatory state by inducing T-cell hyporesponsiveness in a SARS-CoV-2-specific manner using tolerogenic dendritic cells (tolDC). In vitro-activated SARS-CoV-2-specific T cells were isolated and stimulated with SARS-CoV-2 peptide-loaded monocyte-derived tolDC or with SARS-CoV-2 peptide-loaded conventional (conv) DC. We demonstrate a significant decrease in the number of interferon (IFN)-γ spot-forming cells when SARS-CoV-2-specific T cells were stimulated with tolDC as compared to stimulation with convDC. Importantly, this IFN-γ downmodulation in SARS-CoV-2-specific T cells was antigen-specific, since T cells retain their capacity to respond to an unrelated antigen and are not mediated by T cell deletion. Altogether, we have demonstrated that SARS-CoV-2 peptide-pulsed tolDC induces SARS-CoV-2-specific T cell hyporesponsiveness in an antigen-specific manner as compared to stimulation with SARS-CoV-2-specific convDC. These observations underline the clinical potential of tolDC to correct the immunological imbalance in the critically ill.
Collapse
Affiliation(s)
- Mats Van Delen
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium
| | - Ibo Janssens
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium
| | - Amber Dams
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium
| | - Laurence Roosens
- Laboratory of Clinical Biology, Antwerp University Hospital, 2650 Edegem, Belgium
| | - Benson Ogunjimi
- Centre for Health Economics Research & Modeling Infectious Diseases (CHERMID), VAXINFECTIO, University of Antwerp, 2610 Antwerp, Belgium
- Department of Paediatrics, Antwerp University Hospital, 2650 Edegem, Belgium
- Antwerp Center for Translational Immunology and Virology (ACTIV), VAXINFECTIO, University of Antwerp, 2610 Antwerp, Belgium
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, 2020 Antwerp, Belgium
| | - Zwi N. Berneman
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium
- Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, 2650 Edegem, Belgium
| | - Judith Derdelinckx
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium
- Department of Neurology, Antwerp University Hospital, 2650 Edegem, Belgium
| | - Nathalie Cools
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium
- Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, 2650 Edegem, Belgium
| |
Collapse
|
17
|
Shuai Z, Zheng S, Wang K, Wang J, Leung PSC, Xu B. Reestablish immune tolerance in rheumatoid arthritis. Front Immunol 2022; 13:1012868. [PMID: 36248797 PMCID: PMC9561630 DOI: 10.3389/fimmu.2022.1012868] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic progressive autoimmune disease. Despite the wide use of conventional synthetic, targeted and biologic disease modifying anti-rheumatic drugs (DMARDs) to control its radiological progress, nearly all DMARDs are immunologically non-selective and do not address the underlying immunological mechanisms of RA. Patients with RA often need to take various DMARDs long-term or even lifelong and thus, face increased risks of infection, tumor and other adverse reactions. It is logical to modulate the immune disorders and restore immune balance in patients with RA by restoring immune tolerance. Indeed, approaches based on stem cell transplantation, tolerogenic dendritic cells (tolDCs), and antigen-based tolerogenic vaccination are under active investigation, and some have already transformed from wet bench research to clinical investigation during the last decade. Among them, clinical trials on stem cell therapy, especially mesenchymal stem cells (MSCs) transplantation are most investigated and followed by tolDCs in RA patients. On the other hand, despite active laboratory investigations on the use of RA-specific peptide-/protein-based tolerogenic vaccines for T cell, clinical studies on RA patients are much limited. Overall, the preliminary results of these clinical studies are promising and encouraging, demonstrating their safety and effectiveness in the rebalancing of T cell subsets; particular, the recovery of RA-specific Treg with increasing anti-inflammatory cytokines and reduced proinflammatory cytokines. Future studies should focus on the optimization of transplanted stem cells, the preparation of tolDCs, and tolerogenic vaccines with RA-specific protein or peptide, including their dosage, course, and route of administration with well-coordinated multi-center randomized clinical control researches. With the progress of experimental and clinical studies, generating and restoring RA-specific immune tolerance may bring revolutionary changes to the clinical management of RA in the near future.
Collapse
Affiliation(s)
- Ziqiang Shuai
- Department of Sports Injury and Arthroscopic Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Shuang Zheng
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Kang Wang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jian Wang
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- *Correspondence: Bin Xu, ; Patrick S. C. Leung, ; Jian Wang,
| | - Patrick S. C. Leung
- Division of Rheumatology/Allergy and Clinical Immunology, University of California, Davis, Davis, CA, United States
- *Correspondence: Bin Xu, ; Patrick S. C. Leung, ; Jian Wang,
| | - Bin Xu
- Department of Sports Injury and Arthroscopic Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- *Correspondence: Bin Xu, ; Patrick S. C. Leung, ; Jian Wang,
| |
Collapse
|
18
|
Xu B, Ling S, Xu X, Liu X, Wang A, Zhou Y, Luo Y, Li W, Yao X. A New Formulation of Probiotics Attenuates Calcipotriol-Induced Dermatitis by Inducing Regulatory Dendritic Cells. Front Immunol 2021; 12:775018. [PMID: 34868040 PMCID: PMC8634942 DOI: 10.3389/fimmu.2021.775018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 10/25/2021] [Indexed: 12/12/2022] Open
Abstract
Atopic dermatitis (AD) is a recurrent chronic inflammatory skin disease affecting up to 30% of the children population, and immuno-regulatory therapy that could modify the course of disease is urgently needed. Probiotics have demonstrated therapeutic effects on AD and could potentially regulate the disease process. However, the efficacy of probiotics for AD is inconsistent among different studies, which is mainly due to the elusive mechanism and different species and (or) strains used. In this study, we designed a mixture of five strains of probiotics (named IW5) and analyzed the effect and mechanism of IW5 on calcipotriol (MC903)-induced AD-like dermatitis. We found that IW5 significantly alleviated skin inflammation of the MC903-induced AD in mice. Administration with IW5 induced increased production of regulatory T cells and regulatory dendritic cells (DCregs) in the mesenteric lymph nodes. We also found that the diversity of the gut microbiota in the mice with MC903-induced dermatitis was increased after IW5 administration, and the level of butyrate in the gut was elevated. In cell culture, butyrate induced the production of DCregs. Our study revealed the therapeutic effects of a newly designed probiotics mixture and uncovered a possible mechanism, providing a foundation for future clinical studies.
Collapse
Affiliation(s)
- Beilei Xu
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Shiqi Ling
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Xiaoqiang Xu
- Department of Bioinformatics, 01life Institute, Shenzhen, China
| | - Xiaochun Liu
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Ao Wang
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Yuan Zhou
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Yang Luo
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Wei Li
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Xu Yao
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| |
Collapse
|
19
|
Retinoic Acid-Containing Liposomes for the Induction of Antigen-Specific Regulatory T Cells as a Treatment for Autoimmune Diseases. Pharmaceutics 2021; 13:pharmaceutics13111949. [PMID: 34834364 PMCID: PMC8620283 DOI: 10.3390/pharmaceutics13111949] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/09/2021] [Accepted: 11/12/2021] [Indexed: 11/17/2022] Open
Abstract
The current treatment of autoimmune and chronic inflammatory diseases entails systemic immune suppression, which is associated with increased susceptibility to infections. To restore immune tolerance and reduce systemic side effects, a targeted approach using tolerogenic dendritic cells (tolDCs) is being explored. tolDCs are characterized by the expression of CD11c, the major histocompatibility complex (MHC)II and low levels of co-stimulatory molecules CD40 and CD86. In this study, tolDCs were generated using a human-proteoglycan-derived peptide (hPG) and all-trans retinoic acid (RA). RA-tolDCs not only display a tolerogenic phenotype but also can induce an antigen-specific regulatory T cell (Treg) response in vitro. However, further analysis showed that RA-tolDCs make up a heterogeneous population of DCs, with only a small proportion being antigen-associated tolDCs. To increase the homogeneity of this population, 1,2-distearoyl-sn-glycero-3-phosphoglycerol (DSPG)-containing liposomes were used to encapsulate the relevant antigen together with RA. These liposomes greatly enhanced the proportion of antigen-associated tolDCs in culture. In addition, in mice, we showed that the liposomal co-delivery of antigen and RA can be a more targeted approach to induce antigen-specific tolerance compared to the injection of RA-tolDCs, and that these liposomes can stimulate the generation of antigen-specific Tregs. This work highlights the importance of the co-delivery of an antigen and immunomodulator to minimize off-target effects and systemic side effects and provides new insights in the use of RA for antigen-specific immunotherapy for autoimmune and chronic inflammatory diseases.
Collapse
|
20
|
Suuring M, Moreau A. Regulatory Macrophages and Tolerogenic Dendritic Cells in Myeloid Regulatory Cell-Based Therapies. Int J Mol Sci 2021; 22:7970. [PMID: 34360736 PMCID: PMC8348814 DOI: 10.3390/ijms22157970] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/22/2021] [Accepted: 07/23/2021] [Indexed: 12/12/2022] Open
Abstract
Myeloid regulatory cell-based therapy has been shown to be a promising cell-based medicinal approach in organ transplantation and for the treatment of autoimmune diseases, such as type 1 diabetes, rheumatoid arthritis, Crohn's disease and multiple sclerosis. Dendritic cells (DCs) are the most efficient antigen-presenting cells and can naturally acquire tolerogenic properties through a variety of differentiation signals and stimuli. Several subtypes of DCs have been generated using additional agents, including vitamin D3, rapamycin and dexamethasone, or immunosuppressive cytokines, such as interleukin-10 (IL-10) and transforming growth factor-beta (TGF-β). These cells have been extensively studied in animals and humans to develop clinical-grade tolerogenic (tol)DCs. Regulatory macrophages (Mregs) are another type of protective myeloid cell that provide a tolerogenic environment, and have mainly been studied within the context of research on organ transplantation. This review aims to thoroughly describe the ex vivo generation of tolDCs and Mregs, their mechanism of action, as well as their therapeutic application and assessment in human clinical trials.
Collapse
Affiliation(s)
| | - Aurélie Moreau
- Centre de Recherche en Transplantation et Immunologie—UMR1064, INSERM—ITUN, Nantes Université, CHU Nantes, 44000 Nantes, France;
| |
Collapse
|
21
|
Ness S, Lin S, Gordon JR. Regulatory Dendritic Cells, T Cell Tolerance, and Dendritic Cell Therapy for Immunologic Disease. Front Immunol 2021; 12:633436. [PMID: 33777019 PMCID: PMC7988082 DOI: 10.3389/fimmu.2021.633436] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 01/18/2021] [Indexed: 12/12/2022] Open
Abstract
Dendritic cells (DC) are antigen-presenting cells that can communicate with T cells both directly and indirectly, regulating our adaptive immune responses against environmental and self-antigens. Under some microenvironmental conditions DC develop into anti-inflammatory cells which can induce immunologic tolerance. A substantial body of literature has confirmed that in such settings regulatory DC (DCreg) induce T cell tolerance by suppression of effector T cells as well as by induction of regulatory T cells (Treg). Many in vitro studies have been undertaken with human DCreg which, as a surrogate marker of antigen-specific tolerogenic potential, only poorly activate allogeneic T cell responses. Fewer studies have addressed the abilities of, or mechanisms by which these human DCreg suppress autologous effector T cell responses and induce infectious tolerance-promoting Treg responses. Moreover, the agents and properties that render DC as tolerogenic are many and varied, as are the cells’ relative regulatory activities and mechanisms of action. Herein we review the most current human and, where gaps exist, murine DCreg literature that addresses the cellular and molecular biology of these cells. We also address the clinical relevance of human DCreg, highlighting the outcomes of pre-clinical mouse and non-human primate studies and early phase clinical trials that have been undertaken, as well as the impact of innate immune receptors and symbiotic microbial signaling on the immunobiology of DCreg.
Collapse
Affiliation(s)
- Sara Ness
- Department of Medicine, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Shiming Lin
- Department of Medicine, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - John R Gordon
- Department of Medicine, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada.,Division of Respirology, Critical Care and Sleep Medicine, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
22
|
Page A, Fusil F, Cosset FL. Antigen-specific tolerance approach for rheumatoid arthritis: Past, present and future. Joint Bone Spine 2021; 88:105164. [PMID: 33618000 DOI: 10.1016/j.jbspin.2021.105164] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 02/02/2021] [Indexed: 02/06/2023]
Abstract
Rheumatoid arthritis is a chronic systemic autoimmune disease, affecting mainly the joints. It is caused by an adaptive immune reaction against self-antigens, leading to the over production of inflammatory cytokines and autoantibodies, mainly mediated by autoreactive CD4+ T cells and pathological B cell clones. The treatment options currently available rely on palliative global immunosuppression and do not restore tolerance to self-components. Here, we review antigen-specific tolerance approaches that have been developed to inhibit or delete autoreactive clones, while maintaining a potent immune system for rheumatoid arthritis. The first attempts relied on the oral ingestion of self-reactive peptides, with lukewarm results in human clinical trials. To enhance treatment efficacy, self-peptides have been engineered and combined with immunosuppressive molecules. In addition, several routes of delivery have been tested, in particular, nanoparticles carrying self-antigens and immunomodulatory molecules. More recently, transfer of immune cells, such as tolerogenic dendritic cells or regulatory T cells, has been considered to restore tolerance. Although promising results have been obtained in mouse models, the translation to humans remains highly challenging, mainly because the disease is already well developed when treatments start and because patient's specific self-antigens are often unknown. Nevertheless, these approaches hold great promises for long-term RA treatment.
Collapse
Affiliation(s)
- Audrey Page
- CIRI - Centre international de recherche en infectiologie, Université de Lyon, Université Claude-Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, 46, allée d'Italie, 69007 Lyon, France
| | - Floriane Fusil
- CIRI - Centre international de recherche en infectiologie, Université de Lyon, Université Claude-Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, 46, allée d'Italie, 69007 Lyon, France
| | - François-Loïc Cosset
- CIRI - Centre international de recherche en infectiologie, Université de Lyon, Université Claude-Bernard Lyon 1, Inserm, U1111, CNRS, UMR5308, ENS Lyon, 46, allée d'Italie, 69007 Lyon, France.
| |
Collapse
|
23
|
Bassaganya-Riera J, Berry EM, Blaak EE, Burlingame B, le Coutre J, van Eden W, El-Sohemy A, German JB, Knorr D, Lacroix C, Muscaritoli M, Nieman DC, Rychlik M, Scholey A, Serafini M. Goals in Nutrition Science 2020-2025. Front Nutr 2021; 7:606378. [PMID: 33665201 PMCID: PMC7923694 DOI: 10.3389/fnut.2020.606378] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 11/26/2020] [Indexed: 12/12/2022] Open
Abstract
Five years ago, with the editorial board of Frontiers in Nutrition, we took a leap of faith to outline the Goals for Nutrition Science - the way we see it (1). Now, in 2020, we can put ourselves to the test and take a look back. Without a doubt we got it right with several of the key directions. To name a few, Sustainable Development Goals (SDGs) for Food and Nutrition are part of the global public agenda, and the SDGs contribute to the structuring of international science and research. Nutritional Science has become a critical element in strengthening work on the SDGs, and the development of appropriate methodologies is built on the groundwork of acquiring and analyzing big datasets. Investigation of the Human Microbiome is providing novel insight on the interrelationship between nutrition, the immune system and disease. Finally, with an advanced definition of the gut-brain-axis we are getting a glimpse into the potential for Nutrition and Brain Health. Various milestones have been achieved, and any look into the future will have to consider the lessons learned from Covid-19 and the sobering awareness about the frailty of our food systems in ensuring global food security. With a view into the coming 5 years from 2020 to 2025, the editorial board has taken a slightly different approach as compared to the previous Goals article. A mind map has been created to outline the key topics in nutrition science. Not surprisingly, when looking ahead, the majority of scientific investigation required will be in the areas of health and sustainability. Johannes le Coutre, Field Chief Editor, Frontiers in Nutrition.
Collapse
Affiliation(s)
- Josep Bassaganya-Riera
- Nutritional Immunology and Molecular Medicine Laboratory (NIMML) Institute, Blacksburg, VA, United States
| | - Elliot M Berry
- Braun School of Public Health, Hebrew University - Hadassah Medical School, Jerusalem, Israel
| | - Ellen E Blaak
- Department of Human Biology, Maastricht University, Maastricht, Netherlands
| | | | - Johannes le Coutre
- School of Chemical Engineering, University of New South Wales, Sydney, NSW, Australia
| | - Willem van Eden
- Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, Netherlands
| | - Ahmed El-Sohemy
- Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada
| | - J Bruce German
- Department of Food Science and Technology, University of California, Davis, Davis, CA, United States
| | - Dietrich Knorr
- Institute of Food Technology and Chemistry, Technische Universität Berlin, Berlin, Germany
| | - Christophe Lacroix
- Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | - Maurizio Muscaritoli
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - David C Nieman
- Human Performance Laboratory, Department of Biology, Appalachian State University, Kannapolis, NC, United States
| | - Michael Rychlik
- Technical University of Munich, Analytical Food Chemistry, Freising, Germany
| | - Andrew Scholey
- Centre for Human Psychopharmacology, Swinburne University, Melbourne, VIC, Australia
| | - Mauro Serafini
- Functional Food and Metabolic Stress Prevention Laboratory, Faculty of Biosciences and Technologies for Agriculture, Food and Environment, University of Teramo, Teramo, Italy
| |
Collapse
|
24
|
Morante-Palacios O, Fondelli F, Ballestar E, Martínez-Cáceres EM. Tolerogenic Dendritic Cells in Autoimmunity and Inflammatory Diseases. Trends Immunol 2020; 42:59-75. [PMID: 33293219 DOI: 10.1016/j.it.2020.11.001] [Citation(s) in RCA: 136] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 11/06/2020] [Accepted: 11/08/2020] [Indexed: 12/11/2022]
Abstract
Dendritic cells (DCs), the most efficient antigen-presenting cells, are necessary for the effective activation of naïve T cells. DCs can also acquire tolerogenic functions in vivo and in vitro in response to various stimuli, including interleukin (IL)-10, transforming growth factor (TGF)-β, vitamin D3, corticosteroids, and rapamycin. In this review, we provide a wide perspective on the regulatory mechanisms, including crosstalk with other cell types, downstream signaling pathways, transcription factors, and epigenetics, underlying the acquisition of tolerogenesis by DCs, with a special focus on human studies. Finally, we present clinical assays targeting, or based on, tolerogenic DCs in inflammatory diseases. Our discussion provides a useful resource for better understanding the biology of tolerogenic DCs and their manipulation to improve the immunological fitness of patients with certain inflammatory conditions.
Collapse
Affiliation(s)
- Octavio Morante-Palacios
- Epigenetics and Immune Disease Group, Josep Carreras Research Institute (IJC), 08916 Badalona, Barcelona, Spain; Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Barcelona, Spain
| | - Federico Fondelli
- Division of Immunology, Germans Trias i Pujol Hospital, LCMN, Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Barcelona, Spain; Department of Cell Biology, Physiology, Immunology, Autonomous University of Barcelona, 08193 Bellaterra, Barcelona, Spain
| | - Esteban Ballestar
- Epigenetics and Immune Disease Group, Josep Carreras Research Institute (IJC), 08916 Badalona, Barcelona, Spain; Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Barcelona, Spain.
| | - Eva M Martínez-Cáceres
- Division of Immunology, Germans Trias i Pujol Hospital, LCMN, Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Barcelona, Spain; Department of Cell Biology, Physiology, Immunology, Autonomous University of Barcelona, 08193 Bellaterra, Barcelona, Spain.
| |
Collapse
|
25
|
Edilova MI, Akram A, Abdul-Sater AA. Innate immunity drives pathogenesis of rheumatoid arthritis. Biomed J 2020; 44:172-182. [PMID: 32798211 PMCID: PMC8178572 DOI: 10.1016/j.bj.2020.06.010] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 06/29/2020] [Indexed: 02/06/2023] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease affecting ∼1% of the general population. This disease is characterized by persistent articular inflammation and joint damage driven by the proliferating synovial tissue fibroblasts as well as neutrophil, monocyte and lymphocyte trafficking into the synovium. The factors leading to RA pathogenesis remain poorly elucidated although genetic and environmental factors have been proposed to be the main contributors to RA. The majority of the early studies focused on the role of lymphocytes and adaptive immune responses in RA. However, in the past two decades, emerging studies showed that the innate immune system plays a critical role in the onset and progression of RA pathogenesis. Various innate immune cells including monocytes, macrophages and dendritic cells are involved in inflammatory responses seen in RA patients as well as in driving the activation of the adaptive immune system, which plays a major role in the later stages of the disease. Here we focus the discussion on the role of different innate immune cells and components in initiation and progression of RA. New therapeutic approaches targeting different inflammatory pathways and innate immune cells will be highlighted here. Recent emergence and the significant roles of innate lymphoid cells and inflammasomes will be also discussed.
Collapse
Affiliation(s)
- Maria I Edilova
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Ali Akram
- School of Kinesiology and Health Science, Muscle Health Research Centre, York University, Toronto, Ontario Canada; The University Health Network, Toronto Western Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Ali A Abdul-Sater
- School of Kinesiology and Health Science, Muscle Health Research Centre, York University, Toronto, Ontario Canada.
| |
Collapse
|