1
|
McBride DA, Wang JS, Johnson WT, Bottini N, Shah NJ. ABCD of IA: A multi-scale agent-based model of T cell activation in inflammatory arthritis. Biomater Sci 2024; 12:2041-2056. [PMID: 38349277 DOI: 10.1039/d3bm01674a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
Biomaterial-based agents have been demonstrated to regulate the function of immune cells in models of autoimmunity. However, the complexity of the kinetics of immune cell activation can present a challenge in optimizing the dose and frequency of administration. Here, we report a model of autoreactive T cell activation, which are key drivers in autoimmune inflammatory joint disease. The model is termed a multi-scale Agent-Based, Cell-Driven model of Inflammatory Arthritis (ABCD of IA). Using kinetic rate equations and statistical theory, ABCD of IA simulated the activation and presentation of autoantigens by dendritic cells, interactions with cognate T cells and subsequent T cell proliferation in the lymph node and IA-affected joints. The results, validated with in vivo data from the T cell driven SKG mouse model, showed that T cell proliferation strongly correlated with the T cell receptor (TCR) affinity distribution (TCR-ad), with a clear transition state from homeostasis to an inflammatory state. T cell proliferation was strongly dependent on the amount of antigen in antigenic stimulus event (ASE) at low concentrations. On the other hand, inflammation driven by Th17-inducing cytokine mediated T cell phenotype commitment was influenced by the initial level of Th17-inducing cytokines independent of the amount of arthritogenic antigen. The introduction of inhibitory artificial antigen presenting cells (iaAPCs), which locally suppress T cell activation, reduced T cell proliferation in a dose-dependent manner. The findings in this work set up a framework based on theory and modeling to simulate personalized therapeutic strategies in IA.
Collapse
Affiliation(s)
- David A McBride
- Department of NanoEngineering and Chemical Engineering Program, University of California, San Diego, La Jolla, CA 92093, USA.
| | - James S Wang
- Department of NanoEngineering and Chemical Engineering Program, University of California, San Diego, La Jolla, CA 92093, USA.
| | - Wade T Johnson
- Department of NanoEngineering and Chemical Engineering Program, University of California, San Diego, La Jolla, CA 92093, USA.
| | - Nunzio Bottini
- Kao Autoimmunity Institute and Division of Rheumatology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Nisarg J Shah
- Department of NanoEngineering and Chemical Engineering Program, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
2
|
Martínez-Ramos S, García S. An update of murine models and their methodologies in immune-mediated joint damage and pain research. Int Immunopharmacol 2024; 128:111440. [PMID: 38176343 DOI: 10.1016/j.intimp.2023.111440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/13/2023] [Accepted: 12/21/2023] [Indexed: 01/06/2024]
Abstract
Murine models have played an indispensable role in the understanding of rheumatic and musculoskeletal disorders (RMD), elucidating the genetic, endocrine and biomechanical pathways involved in joint pathology and associated pain. To date, the available models in RMD can be classified as induced or spontaneous, both incorporating transgenic alternatives that improve specific insights. It is worth noting that the selection of the most appropriate model together with the evaluation of their specific characteristics and technical capabilities are crucial when designing the experiments. Furthermore, it is also imperative to consistently adhere to the ethical standards concerning animal experimentation. Recognizing the inherent limitation that any model can entirely encapsulates the complexity of the pathophysiology of these conditions, the aim of this review is to provide an updated overview on the methodology of current murine models in major arthropathies and their immune-mediated pathways, addressing to basic, translational and pharmacological research in joint damage and pain.
Collapse
Affiliation(s)
- Sara Martínez-Ramos
- Rheumatology & Immuno-mediated Diseases Research Group (IRIDIS), Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain; Rheumatology Department, University Hospital Complex of Vigo, Vigo, Spain.
| | - Samuel García
- Rheumatology & Immuno-mediated Diseases Research Group (IRIDIS), Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain; Rheumatology Department, University Hospital Complex of Vigo, Vigo, Spain
| |
Collapse
|
3
|
Wang S, Zhou Y, Huang J, Li H, Pang H, Niu D, Li G, Wang F, Zhou Z, Liu Z. Advances in experimental models of rheumatoid arthritis. Eur J Immunol 2023; 53:e2249962. [PMID: 36330559 DOI: 10.1002/eji.202249962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 10/16/2022] [Accepted: 11/02/2022] [Indexed: 11/06/2022]
Abstract
Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease characterized by persistent articular inflammation and joint damage. RA was first described over 200 years ago; however, its etiology and pathophysiology remain insufficiently understood. The current treatment of RA is mainly empirical or based on the current understanding of etiology with limited efficacy and/or substantial side effects. Thus, the development of safer and more potent therapeutics, validated and optimized in experimental models, is urgently required. To improve the transition from bench to bedside, researchers must carefully select the appropriate experimental models as well as draw the right conclusions. Here, we summarize the establishment, pathological features, potential mechanisms, advantages, and limitations of the currently available RA models. The aim of the review is to help researchers better understand available RA models; discuss future trends in RA model development, which can help highlight new translational and human-based avenues in RA research.
Collapse
Affiliation(s)
- Siwei Wang
- School of Basic Medicine, Yangtze University, Jingzhou, Hubei Province, China.,Honghu Hospital of Traditional Chinese Medicine, Affiliated Hospital of Yangtze University, Honghu, Hubei Province, China
| | - Yanhua Zhou
- School of Basic Medicine, Yangtze University, Jingzhou, Hubei Province, China.,Honghu Hospital of Traditional Chinese Medicine, Affiliated Hospital of Yangtze University, Honghu, Hubei Province, China
| | - Jiangrong Huang
- School of Basic Medicine, Yangtze University, Jingzhou, Hubei Province, China
| | - Huilin Li
- School of Basic Medicine, Yangtze University, Jingzhou, Hubei Province, China
| | - Huidan Pang
- School of Basic Medicine, Yangtze University, Jingzhou, Hubei Province, China
| | - Dandan Niu
- School of Basic Medicine, Yangtze University, Jingzhou, Hubei Province, China
| | - Guangyao Li
- School of Basic Medicine, Yangtze University, Jingzhou, Hubei Province, China
| | - Fei Wang
- Department of Experiment and Training, Hubei College of Chinese Medicine, Hubei Province, China
| | - Zushan Zhou
- School of Basic Medicine, Yangtze University, Jingzhou, Hubei Province, China.,Honghu Hospital of Traditional Chinese Medicine, Affiliated Hospital of Yangtze University, Honghu, Hubei Province, China
| | - Zhenzhen Liu
- School of Basic Medicine, Yangtze University, Jingzhou, Hubei Province, China
| |
Collapse
|
4
|
Determination of Therapeutic and Safety Effects of Zygophyllum coccineum Extract in Induced Inflammation in Rats. BIOMED RESEARCH INTERNATIONAL 2022; 2022:7513155. [PMID: 35898689 PMCID: PMC9314163 DOI: 10.1155/2022/7513155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 05/28/2022] [Accepted: 06/14/2022] [Indexed: 11/30/2022]
Abstract
Background Z. coccineum is a facultative plant with many medicinal applications. This study examined the anti-inflammatory activity of Zygophyllum coccineum (Z. coccineum) in an arthritis animal model. Materials and Methods Seventy-Six Wistar Albino rats of either sex randomly divided into six groups (12/each). The inflammation model was done using Complete Freund's Adjuvant in albino rats. The anti-inflammatory activities of the extract were estimated at different dose levels (15.6, 31, and 60 mg/kg) as well as upon using methotrexate (MTX) as a standard drug (0.3 mg/kg). Paw volume and arthritis index scores have been tested in all examined animals' treatments. Histological examination of joints was also performed. Flow cytometric studies were done to isolated osteoclasts. Cytokines assay as well as biochemical testing was done in the examined samples. Results. In vitro studies reported an IC50 of 15.6 μg/ml for Z. coccineum extract in lipoxygenase inhibition assay (L.O.X.). Moreover, it could be noticed that isorhamnetin-3-O-glucoside, tribuloside, and 7-acetoxy-4-methyl coumarin were the most common compounds in Z. coccineum extract separated using L.C.–ESI-TOF–M.S. (liquid chromatography-electrospray ionization ion-trap time-of-flight mass spectrometry). Microscopic examinations of synovial tissue and hind limb muscles revealed the effect of different doses of Z. coccineum extract on restoring chondrocytes and muscles structures. Osteoclast size and apoptotic rate examinations revealed the protective effect of Z. coccineum extract on osteoclast. The results upon induction of animals and upon treatment using of MTX significantly increased apoptotic rate of osteoclast compared to control, while using of 15.6 μg/ml. for Z. coccineum extract lead to recover regular apoptotic rate demonstrating the protective effect of the extract. Z. coccineum extract regulated the secretion of proinflammatory and anti-inflammatory cytokines. Biochemical tests indicated the safety of Z. coccineum extract on kidney and liver functions. Conclusion. Z. coccineum extract has efficient and safe anti-inflammatory potential in an induced rat model.
Collapse
|
5
|
Zhao T, Xie Z, Xi Y, Liu L, Li Z, Qin D. How to Model Rheumatoid Arthritis in Animals: From Rodents to Non-Human Primates. Front Immunol 2022; 13:887460. [PMID: 35693791 PMCID: PMC9174425 DOI: 10.3389/fimmu.2022.887460] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 04/28/2022] [Indexed: 02/05/2023] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease influenced by both genetic and environmental factors. At present, rodent models are primarily used to study the pathogenesis and treatment of RA. However, the genetic divergences between rodents and humans determine differences in the development of RA, which makes it necessary to explore the establishment of new models. Compared to rodents, non-human primates (NHPs) are much more closely related to humans in terms of the immune system, metabolic conditions, and genetic make-up. NHPs model provides a powerful tool to study the development of RA and potential complications, as well as preclinical studies in drug development. This review provides a brief overview of the RA animal models, emphasizes the replication methods, pros and cons, as well as evaluates the validity of the rodent and NHPs models.
Collapse
Affiliation(s)
- Ting Zhao
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, China
| | - Zhaohu Xie
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, China
| | - Yujiang Xi
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, China
| | - Li Liu
- Ge Jiu People’s Hospital, Yunnan Honghe Prefecture Central Hospital, Gejiu, China
| | - Zhaofu Li
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, China
| | - Dongdong Qin
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, China
| |
Collapse
|
6
|
Heluany CS, Scharf P, Schneider AH, Donate PB, Dos Reis Pedreira Filho W, de Oliveira TF, Cunha FQ, Farsky SHP. Toxic mechanisms of cigarette smoke and heat-not-burn tobacco vapor inhalation on rheumatoid arthritis. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 809:151097. [PMID: 34695477 DOI: 10.1016/j.scitotenv.2021.151097] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/24/2021] [Accepted: 10/16/2021] [Indexed: 06/13/2023]
Abstract
Tobacco combustion exposure worsens rheumatoid arthritis (RA). Non-combustible tobacco devices, as heat-not-burn tobacco (HNBT), are emerging as harm reduction to smokers by releasing nicotine and lower combustible tobacco products. Nevertheless, HNBT toxicity remains unclear. Hence, here we investigated the impacts of the tobacco combustible product (cigarette smoke; CS) or HNBT vapor exposures on antigen-induced arthritis (AIA) in C57BL/6 mice. Animals were exposed to airflow, HNBT vapor, or CS during 1 h/twice a day, under the Health Canada Intense (HCI) smoking regime, between days 14 to 20 after the first immunization. At day 21, 16 h after the last exposures, mice were i.a. challenged and the AIA effects were evaluated 24 h later. CS- or HNBT-exposed mice presented equivalent blood nicotine levels. CS exposure worsened articular symptoms, pulmonary inflammation, and expression of lung metallothioneins. Nevertheless, CS or HNBT exposures reduced lymphoid organs' cellularity, splenocyte proliferation and IL-2 secretion. Additional in vitro CS or HNBT exposures confirmed the harmful effects on splenocytes, which were partially mediated by the activation of nicotine/α7nAchR pathway. Associated, data demonstrate the toxic mechanisms of CS or HNBT inhalation at HCI regime on RA, and highlight that further investigations are fundamental to assure the toxicity of emerging tobacco products on the immune system during specific challenges.
Collapse
Affiliation(s)
- Cintia Scucuglia Heluany
- Department of Clinical & Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, SP, Brazil
| | - Pablo Scharf
- Department of Clinical & Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, SP, Brazil
| | | | - Paula Barbim Donate
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Brazil
| | | | - Tiago Franco de Oliveira
- Department of Pharmacosciences, Federal University of Health Sciences of Porto Alegre, Porto Alegre, RS, Brazil
| | - Fernando Queiroz Cunha
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Brazil
| | - Sandra Helena Poliselli Farsky
- Department of Clinical & Toxicological Analyses, School of Pharmaceutical Sciences, University of Sao Paulo, SP, Brazil.
| |
Collapse
|
7
|
Chang MH, Levescot A, Nelson-Maney N, Blaustein RB, Winden KD, Morris A, Wactor A, Balu S, Grieshaber-Bouyer R, Wei K, Henderson LA, Iwakura Y, Clark RA, Rao DA, Fuhlbrigge RC, Nigrovic PA. Arthritis flares mediated by tissue-resident memory T cells in the joint. Cell Rep 2021; 37:109902. [PMID: 34706228 DOI: 10.1016/j.celrep.2021.109902] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 08/20/2021] [Accepted: 10/05/2021] [Indexed: 11/17/2022] Open
Abstract
Rheumatoid arthritis is a systemic autoimmune disease, but disease flares typically affect only a subset of joints, distributed in a distinctive pattern for each patient. Pursuing this intriguing pattern, we show that arthritis recurrence is mediated by long-lived synovial resident memory T cells (TRM). In three murine models, CD8+ cells bearing TRM markers remain in previously inflamed joints during remission. These cells are bona fide TRM, exhibiting a failure to migrate between joints, preferential uptake of fatty acids, and long-term residency. Disease flares result from TRM activation by antigen, leading to CCL5-mediated recruitment of circulating effector cells. Correspondingly, TRM depletion ameliorates recurrence in a site-specific manner. Human rheumatoid arthritis joint tissues contain a comparable CD8+-predominant TRM population, which is most evident in late-stage leukocyte-poor synovium, exhibiting limited T cell receptor diversity and a pro-inflammatory transcriptomic signature. Together, these findings establish synovial TRM as a targetable mediator of disease chronicity in autoimmune arthritis.
Collapse
Affiliation(s)
- Margaret H Chang
- Division of Immunology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Anaïs Levescot
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Nathan Nelson-Maney
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Rachel B Blaustein
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Kellen D Winden
- Department of Neurology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Allyn Morris
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Alexandra Wactor
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Spoorthi Balu
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Ricardo Grieshaber-Bouyer
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Kevin Wei
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Lauren A Henderson
- Division of Immunology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Yoichiro Iwakura
- Center for Experimental Animal Models, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba 278-0022, Japan
| | - Rachael A Clark
- Department of Dermatology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Deepak A Rao
- Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Robert C Fuhlbrigge
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, USA.
| | - Peter A Nigrovic
- Division of Immunology, Boston Children's Hospital, Boston, MA 02115, USA; Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
8
|
Dietary Derived Propionate Regulates Pathogenic Fibroblast Function and Ameliorates Experimental Arthritis and Inflammatory Tissue Priming. Nutrients 2021; 13:nu13051643. [PMID: 34068191 PMCID: PMC8152983 DOI: 10.3390/nu13051643] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/04/2021] [Accepted: 05/10/2021] [Indexed: 12/15/2022] Open
Abstract
Short-chain fatty acids are gut-bacteria-derived metabolites that execute important regulatory functions on adaptive immune responses, yet their influence on inflammation driven by innate immunity remains understudied. Here, we show that propionate treatment in drinking water or upon local application into the joint reduced experimental arthritis and lowered inflammatory tissue priming mediated by synovial fibroblasts. On a cellular level, incubation of synovial fibroblasts with propionate or a physiological mixture of short-chain fatty acids interfered with production of inflammatory mediators and migration and induced immune-regulatory fibroblast senescence. Our study suggests that propionate mediates its alleviating effect on arthritis by direct abrogation of local arthritogenic fibroblast function.
Collapse
|
9
|
Chen Y, Shao S, Huang J, Gu Y, Cheng Y, Zhu X. Therapeutic Efficacy of a Trichinella Spiralis Paramyosin-Derived Peptide Modified With a Membrane-Targeting Signal in Mice With Antigen-Induced Arthritis. Front Microbiol 2020; 11:608380. [PMID: 33424810 PMCID: PMC7785802 DOI: 10.3389/fmicb.2020.608380] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 11/30/2020] [Indexed: 12/21/2022] Open
Abstract
Helminth-derived molecules have the ability to modulate the host immune system. Our previous study identified a tetradecapeptide derived from Trichinella spiralis paramyosin (Ts-pmy) that could bind to human complement component C9 to inhibit its polymerization, making the peptide a candidate therapeutic agent for complement-related immune disorders. Here, the peptide underwent an N-terminal modification with a membrane-targeting signal (a unique myristoylated peptide) to improve its therapeutic efficacy. We found that the modified peptide had a binding affinity to human C9 that was similar to that of the original peptide, as confirmed by microscale thermophoresis assays. The binding of the modified peptide to human C9 resulted in the inhibition of C9-related complement activation, as reflected by the decreased Zn2+-induced C9 polymerization and the decreased C9-dependent lysis of rabbit erythrocytes. In addition, the original and modified peptides could both bind to recombinant mouse C9 and inhibit the C9-dependent lysis of rabbit erythrocytes in normal mouse serum (NMS), which meant that the peptides could cross the species barrier to inhibit complement activity in mice. Further in vitro and in vivo analyses confirmed that the peptide modification increased the retention time of the peptide. Furthermore, intraarticular injection of the modified peptide markedly ameliorated knee swelling and joint damage in mice with antigen-induced arthritis (AIA), as assessed histologically. These results suggested that the Ts-pmy-derived peptide modified with a membrane-targeting signal was a reasonable candidate therapeutic agent for membrane attack complex (MAC)-related diseases [such as rheumatoid arthritis (RA)] and the study presented a new modification method to improve the potential therapeutic effects of the peptide.
Collapse
Affiliation(s)
- Yi Chen
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Shuai Shao
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Jingjing Huang
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yuan Gu
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yuli Cheng
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xinping Zhu
- Department of Medical Microbiology and Parasitology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
10
|
Ortiz G, Ledesma-Colunga MG, Wu Z, García-Rodrigo JF, Adan N, Martínez de la Escalera G, Clapp C. Vasoinhibin reduces joint inflammation, bone loss, and the angiogenesis and vasopermeability of the pannus in murine antigen-induced arthritis. J Transl Med 2020; 100:1068-1079. [PMID: 32341517 DOI: 10.1038/s41374-020-0432-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 04/07/2020] [Accepted: 04/07/2020] [Indexed: 12/21/2022] Open
Abstract
Increased permeability and growth (angiogenesis) of blood vessels play a key role in joint swelling and pannus formation in inflammatory arthritis, a family of diseases influenced by reproductive hormones. The hormone prolactin (PRL) protects against joint inflammation, pannus formation, and bone destruction in adjuvant-induced arthritis and these effects may involve its proteolytic conversion to vasoinhibin, a PRL fragment that inhibits angiogenesis and vasopermeability. Here, we show that the intra-articular injection of an adeno-associated virus type-2 (AAV2) vector encoding vasoinhibin reduced joint inflammation, the hyperplasia, vascular density, and vasopermeability of the pannus, and the loss of bone in mice subjected to antigen-induced arthritis. In agreement, the AAV2 vasoinhibin vector reduced the expression of proinflammatory cytokines (interleukin-1β, interleukin-6), an endothelial cell marker (platelet endothelial cell-adhesion molecule 1), and proangiogenic molecules [vascular endothelial growth factor (VEGF), VEGF receptor 2, and hypoxia-inducible factor 1α] in the arthritic joint. Also, vasoinhibin reduced the synovial vasopermeability induced by the intra-articular injection of VEGF in healthy mice. Finally, vasoinhibin signals by blocking the phosphorylation/activation of endothelial nitric oxide synthase (eNOS) at Ser1179 and the AAV2 vasoinhibin vector inhibited the enhanced phosphorylation of eNOS Ser1179 in the arthritic joint. We conclude that vasoinhibin reduces joint inflammation and bone loss in arthritis by inhibiting pannus angiogenesis and vasopermeability via the blockage of VEGF-induced eNOS activation. These findings suggest the potential therapeutic benefit of AAV2-mediated vasoinhibin gene delivery in arthritis.
Collapse
Affiliation(s)
- Georgina Ortiz
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Querétaro, 76230, México
| | - Maria G Ledesma-Colunga
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Querétaro, 76230, México
| | - Zhijian Wu
- Ocular Gene Therapy Laboratory, Neurobiology, National Eye Institute, NIH, Bethesda, MD, 20892, USA
| | - Jose F García-Rodrigo
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Querétaro, 76230, México
| | - Norma Adan
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Querétaro, 76230, México
| | | | - Carmen Clapp
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, Querétaro, 76230, México.
| |
Collapse
|
11
|
Royzman D, Andreev D, Stich L, Rauh M, Bäuerle T, Ellmann S, Boon L, Kindermann M, Peckert K, Bozec A, Schett G, Steinkasserer A, Zinser E. Soluble CD83 Triggers Resolution of Arthritis and Sustained Inflammation Control in IDO Dependent Manner. Front Immunol 2019; 10:633. [PMID: 31001257 PMCID: PMC6455294 DOI: 10.3389/fimmu.2019.00633] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 03/08/2019] [Indexed: 12/29/2022] Open
Abstract
Interference with autoimmune-mediated cytokine production is a key yet poorly developed approach to treat autoimmune and inflammatory diseases such as rheumatoid arthritis. Herein, we show that soluble CD83 (sCD83) enhances the resolution of autoimmune antigen-induced arthritis (AIA) by strongly reducing the expression levels of cytokines such as IL-17A, IFNγ, IL-6, and TNFα within the joints. Noteworthy, also the expression of RANKL, osteoclast differentiation, and joint destruction was significantly inhibited by sCD83. In addition, osteoclasts which were cultured in the presence of synovial T cells, derived from sCD83 treated AIA mice, showed a strongly reduced number of multinuclear large osteoclasts compared to mock controls. Enhanced resolution of arthritis by sCD83 was mechanistically based on IDO, since inhibition of IDO by 1-methyltryptophan completely abrogated sCD83 effects on AIA. Blocking experiments, using anti-TGF-β antibodies further revealed that also TGF-β is mechanistically involved in the sCD83 induced reduction of bone destruction and cartilage damage as well as enhanced resolution of inflammation. Resolution of arthritis was associated with increased numbers of regulatory T cells, which are induced in a sCD83-IDO-TGF-β dependent manner. Taken together, sCD83 represents an interesting approach for downregulating cytokine production, inducing regulatory T cells and inducing resolution of autoimmune arthritis.
Collapse
MESH Headings
- Animals
- Antibodies, Blocking/pharmacology
- Antigens, CD/immunology
- Arthritis, Experimental/drug therapy
- Arthritis, Experimental/immunology
- Arthritis, Experimental/pathology
- Cytokines/immunology
- Female
- Immunoglobulins/immunology
- Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors
- Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology
- Inflammation/drug therapy
- Inflammation/immunology
- Inflammation/pathology
- Joints/immunology
- Joints/pathology
- Membrane Glycoproteins/immunology
- Mice
- Signal Transduction/drug effects
- Signal Transduction/immunology
- Solubility
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/pathology
- Transforming Growth Factor beta/antagonists & inhibitors
- Transforming Growth Factor beta/immunology
- Tryptophan/analogs & derivatives
- Tryptophan/pharmacology
- CD83 Antigen
Collapse
Affiliation(s)
- Dmytro Royzman
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Darja Andreev
- Department of Internal Medicine 3, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Lena Stich
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Manfred Rauh
- Department of Pediatrics and Adolescent Medicine, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Tobias Bäuerle
- Institute of Radiology, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Stephan Ellmann
- Institute of Radiology, Universitätsklinikum Erlangen, Erlangen, Germany
| | | | - Markus Kindermann
- Department of Internal Medicine 1, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Katrin Peckert
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Aline Bozec
- Department of Internal Medicine 3, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Georg Schett
- Department of Internal Medicine 3, Universitätsklinikum Erlangen, Erlangen, Germany
| | | | - Elisabeth Zinser
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
12
|
Chenna Narendra S, Chalise JP, Biggs S, Kalinke U, Magnusson M. Regulatory T-Cells Mediate IFN-α-Induced Resistance against Antigen-Induced Arthritis. Front Immunol 2018. [PMID: 29515584 PMCID: PMC5826073 DOI: 10.3389/fimmu.2018.00285] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Objective CD4+FoxP3+CD25+ regulatory T-cells (Tregs) are important for preventing tissue destruction. Here, we investigate the role of Tregs for protection against experimental arthritis by IFN-α. Methods Arthritis was triggered by intra-articular injection of methylated bovine serum albumin (mBSA) in wild-type mice, Foxp3DTReGFP+/− mice [allowing selective depletion of Tregs by diphtheria toxin (DT)] and CD4-Cre+/− IFNA1R flox/flox mice (devoid of IFNAR signaling in T-cells) earlier immunized with mBSA, with or without treatment with IFN-α or the indoleamine 2,3-dioxygenase (IDO)-metabolite kynurenine. Tregs were depleted in DT-treated Foxp3DTReGFP+/− mice and enumerated by FoxP3 staining. Suppressive capacity of FACS-sorted CD25+highCD4+ Tregs was tested in vivo by adoptive transfer and ex vivo in cocultures with antigen-stimulated CFSE-stained T-responder (CD25−CD4+) cells. IDO was inhibited by 1-methyl tryptophan. Results Both control mice and mice devoid of IFNAR-signaling in T helper cells were protected from arthritis by IFN-α. Depletion of Tregs in the arthritis phase, but not at immunization, abolished the protective effect of IFN-α and kynurenine against arthritis. IFN-α increased the number of Tregs in ex vivo cultures upon antigen recall stimulation but not in naïve cells. IFN-α also increased the suppressive capacity of Tregs against mBSA-induced T-responder cell proliferation ex vivo and against arthritis when adoptively transferred. The increased suppressive activity against proliferation conferred by IFN-α was clearly reduced by in vivo inhibition of IDO at immunization, which also abolished the protective effect of IFN-α against arthritis. Conclusion By activating IDO during antigen sensitization, IFN-α activates Tregs, which prevent arthritis triggered by antigen rechallenge. This is one way by which IFN-α suppresses inflammation.
Collapse
Affiliation(s)
- Sudeep Chenna Narendra
- Division of Rheumatology, Autoimmunity and Immune Regulation, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | | | - Sophie Biggs
- Division of Rheumatology, Autoimmunity and Immune Regulation, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Ulrich Kalinke
- Twincore, Zentrum für Experimentelle und Klinische Infektionsforschung, Hannover, Germany
| | - Mattias Magnusson
- Division of Rheumatology, Autoimmunity and Immune Regulation, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| |
Collapse
|
13
|
Abstract
In vivo mouse models of inflammatory arthritis are extensively used to investigate pathogenic mechanisms governing inflammation-driven joint damage. Two commonly utilized models include collagen-induced arthritis (CIA) and methylated bovine serum albumin (mBSA) antigen-induced arthritis (AIA). These offer unique advantages for modeling different aspects of human disease. CIA involves breach of immunological tolerance resulting in systemic autoantibody-driven arthritis, while AIA results in local resolving inflammatory flares and articular T cell-mediated damage. Despite limitations that apply to all animal models of human disease, CIA and AIA have been instrumental in identifying pathogenic mediators, immune cell subsets and stromal cell responses that determine disease onset, progression, and severity. Moreover, these models have enabled investigation of disease phases not easily studied in patients and have served as testing beds for novel biological therapies, including cytokine blockers and small molecule inhibitors of intracellular signaling that have revolutionized rheumatoid arthritis treatment.
Collapse
MESH Headings
- Animals
- Antigens/adverse effects
- Arthritis, Experimental/etiology
- Arthritis, Experimental/metabolism
- Arthritis, Experimental/pathology
- Arthritis, Rheumatoid/etiology
- Arthritis, Rheumatoid/metabolism
- Arthritis, Rheumatoid/pathology
- Cytokines/metabolism
- Disease Models, Animal
- Humans
- Inflammation/etiology
- Inflammation/metabolism
- Inflammation/pathology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Inbred DBA
- Serum Albumin, Bovine/administration & dosage
- Serum Albumin, Bovine/immunology
Collapse
Affiliation(s)
- Gareth W Jones
- Division of Infection and Immunity, Systems Immunity University Research Institute, School of Medicine, Cardiff University, Cardiff, Wales, UK.
| | - David G Hill
- Division of Infection and Immunity, Systems Immunity University Research Institute, School of Medicine, Cardiff University, Cardiff, Wales, UK
| | - Katie Sime
- Division of Infection and Immunity, Systems Immunity University Research Institute, School of Medicine, Cardiff University, Cardiff, Wales, UK
| | - Anwen S Williams
- Division of Infection and Immunity, Systems Immunity University Research Institute, School of Medicine, Cardiff University, Cardiff, Wales, UK
| |
Collapse
|
14
|
Periploca forrestii saponin ameliorates CIA via suppressing proinflammatory cytokines and nuclear factor kappa-B pathways. PLoS One 2017; 12:e0176672. [PMID: 28463993 PMCID: PMC5412996 DOI: 10.1371/journal.pone.0176672] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 04/16/2017] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE Periploca forrestii Schltr has been used as a Chinese folk medicine for the treatment of rheumatism, arthralgia and fractures. However, the anti-arthritic activity of Periploca forrestii saponin (PFS) and the active compound has still not been revealed. This study aimed to investigate the protective effects and mechanisms of PFS on collagen type II (CII) collagen-induced arthritis (CIA) mice. We sought to investigate whether PFS and Periplocin could regulate osteoclastogenesis, and if so, further investigation on its mechanism of action. METHODS Arthritis was induced in female BALB/c mice by CIA method. PFS was administered at a dose of 50 mg/kg body weight once daily for five weeks. The effects of treatment in mice were assessed by histological and biochemical evaluation in sera and paws. Anti-osteoclastogenic action of PFS and Periplocin was identified using an osteoclast formation model induced by RANKL. RESULTS PFS ameliorated paw erythema and swelling, inhibited bone erosion in ankle joint histopathological examination. PFS treatment resulted in decreased IgG2a, and increased IgG1 levels in the serum of CIA mice. Decreased TNF-α, and increased interleukin (IL)-4 and IL-22 levels were also found in PFS-treated mice. PFS inhibited the I-κBα phosphorylation, blocked nuclear factor (NF)-κB/p65 phosphorylation and abrogated AP-1/c-Fos activity. PFS downregulated toll-like receptor (TLR) 4, STAT3 and MMP-9 expression in CIA mice and RANKL-induced osteoclastogenesis. PFS and Periplocin inhibited RANKL-induced osteoclast formation in a dose dependent manner within nongrowth inhibitory concentration, and PFS decreased osteoclastogenesis-related marker expression, including cathepsin K and MMP-9. CONCLUSION This study revealed that the protective mechanism of PFS on CIA was associated with regulatory effects on proinflammatory factors and further on the crosstalk between NF-κB and c-Fos/AP-1 in vivo and in vitro. Therefore, PFS is a promising therapeutic alternative for the treatment of RA, evidencing the need to conduct further studies that can identify their active components in treating and preventing RA.
Collapse
|
15
|
Chalise JP, Pallotta MT, Narendra SC, Carlsson B, Iacono A, Namale J, Boon L, Grohmann U, Magnusson M. IDO1 and TGF-β Mediate Protective Effects of IFN-α in Antigen-Induced Arthritis. THE JOURNAL OF IMMUNOLOGY 2016; 197:3142-3151. [PMID: 27647832 DOI: 10.4049/jimmunol.1502125] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 08/21/2016] [Indexed: 01/06/2023]
Abstract
IFN-α prevents Ag-induced arthritis (AIA), and in this study we investigated the role of IDO1 and TGF-β signaling for this anti-inflammatory property of IFN-α. Arthritis was induced by methylated BSA (mBSA) in mBSA-sensitized wild-type (WT), Ido1-/-, or Ifnar-/- mice, treated or not with IFN-α or the IDO1 product kynurenine (Kyn). Enzymatic IDO1 activity, TGF-β, and plasmacytoid dendritic cells (pDC) were neutralized by 1-methyltryptophan and Abs against TGF-β and pDC, respectively. IDO1 expression was determined by RT-PCR, Western blot, and FACS, and enzymatic activity by HPLC. Proliferation was measured by 3H-thymidine incorporation and TGF-β by RT-PCR and ELISA. WT but not Ido1-/- mice were protected from AIA by IFN-α, and Kyn, the main IDO1 product, also prevented AIA, both in WT and Ifnar-/- mice. Protective treatment with IFN-α increased the expression of IDO1 in pDC during AIA, and Ab-mediated depletion of pDC, either during mBSA sensitization or after triggering of arthritis, completely abrogated the protective effect of IFN-α. IFN-α treatment also increased the enzymatic IDO1 activity (Kyn/tryptophan ratio), which in turn activated production of TGF-β. Neutralization of enzymatic IDO1 activity or TGF-β signaling blocked the protective effect of IFN-α against AIA, but only during sensitization and not after triggering of arthritis. Likewise, inhibition of the IDO1 enzymatic activity in the sensitization phase, but not after triggering of arthritis, subdued the IFN-α-induced inhibition of mBSA-induced proliferation. In conclusion, presence of IFN-α at Ag sensitization activates an IDO1/TGF-β-dependent anti-inflammatory program that upon antigenic rechallenge prevents inflammation via pDC.
Collapse
Affiliation(s)
- Jaya Prakash Chalise
- Division of Rheumatology, Autoimmunity, and Immune Regulation, Department of Clinical and Experimental Medicine, Linköping University, Linköping 58185, Sweden;
| | | | - Sudeep Chenna Narendra
- Division of Rheumatology, Autoimmunity, and Immune Regulation, Department of Clinical and Experimental Medicine, Linköping University, Linköping 58185, Sweden
| | - Björn Carlsson
- Division of Drug Research, Clinical Pharmacology, Department of Medical and Health Sciences, Linköping University, Linköping 58185, Sweden; and
| | - Alberta Iacono
- Department of Experimental Medicine, University of Perugia, 06100 Perugia, Italy
| | - Joanitah Namale
- Division of Rheumatology, Autoimmunity, and Immune Regulation, Department of Clinical and Experimental Medicine, Linköping University, Linköping 58185, Sweden
| | - Louis Boon
- EPIRUS Biopharmaceuticals Netherlands BV, 3584 CM Utrecht, the Netherlands
| | - Ursula Grohmann
- Department of Experimental Medicine, University of Perugia, 06100 Perugia, Italy
| | - Mattias Magnusson
- Division of Rheumatology, Autoimmunity, and Immune Regulation, Department of Clinical and Experimental Medicine, Linköping University, Linköping 58185, Sweden
| |
Collapse
|
16
|
Abstract
Interferon regulatory factor 5 (IRF5) has been demonstrated as a key transcription factor of the immune system, playing important roles in modulating inflammatory immune responses in numerous cell types including dendritic cells, macrophages, and B cells. As well as driving the expression of type I interferon in antiviral responses, IRF5 is also crucial for driving macrophages toward a proinflammatory phenotype by regulating cytokine and chemokine expression and modulating B-cell maturity and antibody production. This review highlights the functional importance of IRF5 in a disease setting, by discussing polymorphic mutations at the human Irf5 locus that lead to susceptibility to systemic lupus erythematosus, rheumatoid arthritis, and inflammatory bowel disease. In concordance with this, we also discuss lessons in IRF5 functionality learned from murine in vivo models of autoimmune disease and inflammation and hypothesize that modulation of IRF5 activity and expression could provide potential therapeutic benefits in the clinic.
Collapse
Affiliation(s)
- Hayley L Eames
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom.
| | - Alastair L Corbin
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Irina A Udalova
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
17
|
Leukocyte-specific protein 1 regulates T-cell migration in rheumatoid arthritis. Proc Natl Acad Sci U S A 2015; 112:E6535-43. [PMID: 26554018 DOI: 10.1073/pnas.1514152112] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Copy number variations (CNVs) have been implicated in human diseases. However, it remains unclear how they affect immune dysfunction and autoimmune diseases, including rheumatoid arthritis (RA). Here, we identified a novel leukocyte-specific protein 1 (LSP1) deletion variant for RA susceptibility located in 11p15.5. We replicated that the copy number of LSP1 gene is significantly lower in patients with RA, which correlates positively with LSP1 protein expression levels. Differentially expressed genes in Lsp1-deficient primary T cells represent cell motility and immune and cytokine responses. Functional assays demonstrated that LSP1, induced by T-cell receptor activation, negatively regulates T-cell migration by reducing ERK activation in vitro. In mice with T-cell-dependent chronic inflammation, loss of Lsp1 promotes migration of T cells into the target tissues as well as draining lymph nodes, exacerbating disease severity. Moreover, patients with RA show diminished expression of LSP1 in peripheral T cells with increased migratory capacity, suggesting that the defect in LSP1 signaling lowers the threshold for T-cell activation. To our knowledge, our work is the first to demonstrate how CNVs result in immune dysfunction and a disease phenotype. Particularly, our data highlight the importance of LSP1 CNVs and LSP1 insufficiency in the pathogenesis of RA and provide previously unidentified insights into the mechanisms underlying T-cell migration toward the inflamed synovium in RA.
Collapse
|
18
|
Chenna Narendra S, Chalise JP, Magnusson M, Uppugunduri S. Local but Not Systemic Administration of Uridine Prevents Development of Antigen-Induced Arthritis. PLoS One 2015; 10:e0141863. [PMID: 26512984 PMCID: PMC4625961 DOI: 10.1371/journal.pone.0141863] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 10/14/2015] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE Uridine has earlier been show to down modulate inflammation in models of lung inflammation. The aim of this study was to evaluate the anti-inflammatory effect of uridine in arthritis. METHODS Arthritis was induced by intra-articular injection of mBSA in the knee of NMRI mice pre-immunized with mBSA. Uridine was either administered locally by direct injection into the knee joint or systemically. Systemic treatment included repeated injections or implantation of a pellet continuously releasing uridine during the entire experimental procedure. Anti-mBSA specific immune responses were determined by ELISA and cell proliferation and serum cytokine levels were determined by Luminex. Immunohistochemistry was used to identify cells, study expression of cytokines and adhesion molecules in the joint. RESULTS Local administration of 25-100 mg/kg uridine at the time of arthritis onset clearly prevented development of joint inflammation. In contrast, systemic administration of uridine (max 1.5 mg uridine per day) did not prevent development of arthritis. Protection against arthritis by local administration of uridine did not affect the anti-mBSA specific immune response and did not prevent the rise in serum levels of pro-inflammatory cytokines associated with the triggering of arthritis. In contrast, local uridine treatment efficiently inhibited synovial expression of ICAM-1 and CD18, local cytokine production and recruitment of leukocytes to the synovium. CONCLUSION Local, but not systemic administration of uridine efficiently prevented development of antigen-induced arthritis. The protective effect did not involve alteration of systemic immunity to mBSA but clearly involved inhibition of synovial expression of adhesion molecules, decreased TNF and IL-6 production and prevention of leukocyte extravasation. Further, uridine is a small, inexpensive molecule and may thus be a new therapeutic option to treat joint inflammation in RA.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/administration & dosage
- Antigens/immunology
- Arthritis, Experimental/drug therapy
- Arthritis, Experimental/immunology
- Arthritis, Experimental/metabolism
- Arthritis, Experimental/pathology
- CD18 Antigens/metabolism
- Cytokines/blood
- Cytokines/metabolism
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Female
- Immunoglobulin G/immunology
- Immunohistochemistry
- Injections, Intra-Articular
- Intercellular Adhesion Molecule-1/metabolism
- Mice
- Serum Albumin, Bovine/adverse effects
- Serum Albumin, Bovine/immunology
- Synovial Membrane/immunology
- Synovial Membrane/metabolism
- Uridine/administration & dosage
Collapse
Affiliation(s)
- Sudeep Chenna Narendra
- Autoimmunity & Immune Regulation (AIR), Department of Clinical & Experimental Medicine, Linköping University, Linköping, Sweden
- * E-mail:
| | - Jaya Prakash Chalise
- Autoimmunity & Immune Regulation (AIR), Department of Clinical & Experimental Medicine, Linköping University, Linköping, Sweden
| | - Mattias Magnusson
- Autoimmunity & Immune Regulation (AIR), Department of Clinical & Experimental Medicine, Linköping University, Linköping, Sweden
| | - Srinivas Uppugunduri
- Department of Clinical Chemistry and Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| |
Collapse
|
19
|
Huang YF, Lo PC, Yen CL, Nigrovic PA, Chao WC, Wang WZ, Hsu GC, Tsai YS, Shieh CC. Redox Regulation of Pro-IL-1β Processing May Contribute to the Increased Severity of Serum-Induced Arthritis in NOX2-Deficient Mice. Antioxid Redox Signal 2015; 23:973-84. [PMID: 25867281 PMCID: PMC4624247 DOI: 10.1089/ars.2014.6136] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
AIMS To elucidate the role of reactive oxygen species (ROS) in arthritis and to identify targets of arthritis treatment in conditions with different levels of oxidant stress. RESULTS Through establishing an arthritis model by injecting arthritogenic serum into wild-type and NADPH oxidase 2 (NOX2)-deficient mice, we found that arthritis had a neutrophilic infiltrate and was more severe in Ncf1(-/-) mice, a mouse strain lacking the expression of the NCF1/p47(phox) component of NOX2. The levels of interleukin-1β (IL-1β) and IL-6 in inflamed joints were higher in Ncf1(-/-) than in controls. Antagonists of tumor necrosis factor-α (TNFα) and IL-1β were equally effective in suppressing arthritis in wild-type mice, while IL-1β blockade was more effective than TNFα blockade in Ncf1(-/-) mice. A treatment of caspase inhibitor and the combination treatment of a caspase inhibitor and a cathepsin inhibitor, but not a cathepsin inhibitor alone, suppressed arthritic severity in the wild-type mice, while a treatment of cathepsin inhibitor and the combination treatment of a caspase inhibitor and a cathepsin inhibitor, but not a caspase inhibitor alone, were effective in treating Ncf1(-/-) mice. Consistently, cathepsin B was found to proteolytically process pro-IL-1β to its active form and this activity was suppressed by ROS. INNOVATION This novel mechanism of a redox-mediated immune regulation of arthritis through leukocyte-produced ROS is important for devising an optimal treatment for patients with different levels of tissue ROS. CONCLUSION Our results suggest that ROS act as a negative feedback to constrain IL-1β-mediated inflammation, accounting for the more severe arthritis in the absence of NOX2.
Collapse
Affiliation(s)
- Ya-Fang Huang
- 1 Institute of Clinical Medicine, National Cheng Kung University College of Medicine , Tainan, Taiwan
| | - Pei-Chi Lo
- 1 Institute of Clinical Medicine, National Cheng Kung University College of Medicine , Tainan, Taiwan
| | - Chia-Liang Yen
- 2 Institute of Basic Medical Science, National Cheng Kung University College of Medicine , Tainan, Taiwan
| | - Peter Andrija Nigrovic
- 3 Division of Rheumatology, Immunology, and Allergy, Brigham and Women's Hospital , Boston, Massachusetts.,4 Division of Immunology, Boston Children's Hospital , Boston, Massachusetts
| | - Wen-Chen Chao
- 1 Institute of Clinical Medicine, National Cheng Kung University College of Medicine , Tainan, Taiwan .,5 Department of Internal Medicine, Taichung Veteran General Hospital , Chiayi Branch, Chiayi, Taiwan
| | - Wei-Zhi Wang
- 1 Institute of Clinical Medicine, National Cheng Kung University College of Medicine , Tainan, Taiwan
| | - George Chengkang Hsu
- 1 Institute of Clinical Medicine, National Cheng Kung University College of Medicine , Tainan, Taiwan
| | - Yau-Sheng Tsai
- 1 Institute of Clinical Medicine, National Cheng Kung University College of Medicine , Tainan, Taiwan
| | - Chi-Chang Shieh
- 1 Institute of Clinical Medicine, National Cheng Kung University College of Medicine , Tainan, Taiwan .,6 Department of Pediatrics, National Cheng Kung University Hospital , Tainan, Taiwan
| |
Collapse
|
20
|
Süß P, Kalinichenko L, Baum W, Reichel M, Kornhuber J, Loskarn S, Ettle B, Distler JHW, Schett G, Winkler J, Müller CP, Schlachetzki JCM. Hippocampal structure and function are maintained despite severe innate peripheral inflammation. Brain Behav Immun 2015; 49:156-70. [PMID: 26074461 DOI: 10.1016/j.bbi.2015.05.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Revised: 05/15/2015] [Accepted: 05/16/2015] [Indexed: 11/17/2022] Open
Abstract
Chronic peripheral inflammation mediated by cytokines such as TNFα, IL-1β, and IL-6 is associated with psychiatric disorders like depression and anxiety. However, it remains elusive which distinct type of peripheral inflammation triggers neuroinflammation and affects hippocampal plasticity resulting in depressive-like behavior. We hypothesized that chronic peripheral inflammation in the human TNF-α transgenic (TNFtg) mouse model of rheumatoid arthritis spreads into the central nervous system and induces depressive state manifested in specific behavioral pattern and impaired adult hippocampal neurogenesis. TNFtg mice showed severe erosive arthritis with increased IL-1β and IL-6 expression in tarsal joints with highly elevated human TNF-α levels in the serum. Intriguingly, IL-1β and IL-6 mRNA levels were not altered in the hippocampus of TNFtg mice. In contrast to the pronounced monocytosis in joints and spleen of TNFtg mice, signs of hippocampal microgliosis or astrocytosis were lacking. Furthermore, locomotion was impaired, but there was no locomotion-independent depressive behavior in TNFtg mice. Proliferation and maturation of hippocampal neural precursor cells as well as survival of newly generated neurons were preserved in the dentate gyrus of TNFtg mice despite reduced motor activity and peripheral inflammatory signature. We conclude that peripheral inflammation in TNFtg mice is mediated by chronic activation of the innate immune system. However, severe peripheral inflammation, though impairing locomotor activity, does not elicit depressive-like behavior. These structural and functional findings indicate the maintenance of hippocampal immunity, cellular plasticity, and behavior despite peripheral innate inflammation.
Collapse
Affiliation(s)
- Patrick Süß
- Department of Molecular Neurology, Friedrich-Alexander-University Erlangen-Nürnberg, University Hospital Erlangen, Schwabachanlage 6, 91054 Erlangen, Germany
| | - Liubov Kalinichenko
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-University Erlangen-Nürnberg, University Hospital Erlangen, Schwabachanlage 6, 91054 Erlangen, Germany
| | - Wolfgang Baum
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg, University Hospital Erlangen, Ulmenweg 18, 91054 Erlangen, Germany
| | - Martin Reichel
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-University Erlangen-Nürnberg, University Hospital Erlangen, Schwabachanlage 6, 91054 Erlangen, Germany
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-University Erlangen-Nürnberg, University Hospital Erlangen, Schwabachanlage 6, 91054 Erlangen, Germany
| | - Sandra Loskarn
- Department of Molecular Neurology, Friedrich-Alexander-University Erlangen-Nürnberg, University Hospital Erlangen, Schwabachanlage 6, 91054 Erlangen, Germany
| | - Benjamin Ettle
- Department of Molecular Neurology, Friedrich-Alexander-University Erlangen-Nürnberg, University Hospital Erlangen, Schwabachanlage 6, 91054 Erlangen, Germany
| | - Jörg H W Distler
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg, University Hospital Erlangen, Ulmenweg 18, 91054 Erlangen, Germany
| | - Georg Schett
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg, University Hospital Erlangen, Ulmenweg 18, 91054 Erlangen, Germany
| | - Jürgen Winkler
- Department of Molecular Neurology, Friedrich-Alexander-University Erlangen-Nürnberg, University Hospital Erlangen, Schwabachanlage 6, 91054 Erlangen, Germany
| | - Christian P Müller
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-University Erlangen-Nürnberg, University Hospital Erlangen, Schwabachanlage 6, 91054 Erlangen, Germany
| | - Johannes C M Schlachetzki
- Department of Molecular Neurology, Friedrich-Alexander-University Erlangen-Nürnberg, University Hospital Erlangen, Schwabachanlage 6, 91054 Erlangen, Germany.
| |
Collapse
|
21
|
Tiniakou I, Drakos E, Sinatkas V, Van Eck M, Zannis VI, Boumpas D, Verginis P, Kardassis D. High-density lipoprotein attenuates Th1 and th17 autoimmune responses by modulating dendritic cell maturation and function. THE JOURNAL OF IMMUNOLOGY 2015; 194:4676-87. [PMID: 25870241 DOI: 10.4049/jimmunol.1402870] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 03/16/2015] [Indexed: 12/11/2022]
Abstract
Aberrant levels and function of the potent anti-inflammatory high-density lipoprotein (HDL) and accelerated atherosclerosis have been reported in patients with autoimmune inflammatory diseases. Whether HDL affects the development of an autoimmune response remains elusive. In this study, we used apolipoprotein A-I-deficient (apoA-I(-/-)) mice, characterized by diminished circulating HDL levels, to delineate the role of HDL in autoimmunity. ApoA-I(-/-) mice exhibited increased severity of Ag-induced arthritis compared with wild-type mice, and this was associated with elevated Th1 and Th17 cell reactivity in the draining lymph nodes. Furthermore, reconstituted HDL (rHDL) attenuated IFN-γ and IL-17 secretion by Ag-specific T cells upon stimulation of draining lymph nodes in vitro. The suppressive effects of rHDL were mediated through modulation of dendritic cell (DC) function. Specifically, rHDL-treated DCs demonstrated an immature phenotype characterized by downregulated costimulatory molecules, the release of low amounts of proinflammatory cytokines, and failure to promote T cell proliferation in vitro. The mechanism of action involved the inhibition of NF-κB nuclear translocation and the decrease of Myd88 mRNA levels by rHDL. Finally, modulation of DC function by rHDL was critically dependent on the presence of scavenger receptor class B type I and ATP Binding Cassette Transporter A1, but not the ATP Binding Cassette Transporter G1. These findings reveal a novel role of HDL in the regulation of adaptive inflammatory responses through suppression of DC function that could be exploited therapeutically in autoimmune inflammatory diseases.
Collapse
Affiliation(s)
- Ioanna Tiniakou
- Laboratory of Biochemistry, Department of Basic Sciences, University of Crete Medical School, 71003 Heraklion, Crete, Greece; Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, 70013 Heraklion, Crete, Greece
| | - Elias Drakos
- Department of Pathology, University of Crete Medical School, 71003 Heraklion, Crete, Greece
| | - Vaios Sinatkas
- Department of Pathology, University of Crete Medical School, 71003 Heraklion, Crete, Greece
| | - Miranda Van Eck
- Division of Biopharmaceutics, Leiden Academic Centre for Drug Research, 2333 Leiden, the Netherlands
| | - Vassilis I Zannis
- Laboratory of Biochemistry, Department of Basic Sciences, University of Crete Medical School, 71003 Heraklion, Crete, Greece; Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118; and
| | - Dimitrios Boumpas
- Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece
| | | | - Dimitris Kardassis
- Laboratory of Biochemistry, Department of Basic Sciences, University of Crete Medical School, 71003 Heraklion, Crete, Greece; Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, 70013 Heraklion, Crete, Greece;
| |
Collapse
|
22
|
Engdahl C, Börjesson AE, Forsman HF, Andersson A, Stubelius A, Krust A, Chambon P, Islander U, Ohlsson C, Carlsten H, Lagerquist MK. The role of total and cartilage-specific estrogen receptor alpha expression for the ameliorating effect of estrogen treatment on arthritis. Arthritis Res Ther 2014; 16:R150. [PMID: 25028072 PMCID: PMC4226038 DOI: 10.1186/ar4612] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Accepted: 07/01/2014] [Indexed: 12/19/2022] Open
Abstract
INTRODUCTION Estrogen (E2) delays onset and decreases severity of experimental arthritis. The aim of this study was to investigate the importance of total estrogen receptor alpha (ERα) expression and cartilage-specific ERα expression in genetically modified mice for the ameliorating effect of estrogen treatment in experimental arthritis. METHODS Mice with total (total ERα-/-) or cartilage-specific (Col2α1-ERα-/-) inactivation of ERα and wild-type (WT) littermates were ovariectomized, treated with E2 or placebo, and induced with antigen-induced arthritis (AIA). At termination, knees were collected for histology, synovial and splenic cells were investigated by using flow cytometry, and splenic cells were subjected to a T-cell proliferation assay. RESULTS E2 decreased synovitis and joint destruction in WT mice. Amelioration of arthritis was associated with decreased frequencies of inflammatory cells in synovial tissue and decreased splenic T-cell proliferation. E2 did not affect synovitis or joint destruction in total ERα-/- mice. In Col2α1-ERα-/- mice, E2 protected against joint destruction to a similar extent as in WT mice. In contrast, E2 did not significantly ameliorate synovitis in Col2α1-ERα-/- mice. CONCLUSIONS Treatment with E2 ameliorates both synovitis and joint destruction in ovariectomized mice with AIA via ERα. This decreased severity in arthritis is associated with decreased synovial inflammatory cell frequencies and reduced splenic T-cell proliferation. ERα expression in cartilage is not required for estrogenic amelioration of joint destruction. However, our data indicate that ERα expression in cartilage is involved in estrogenic effects on synovitis, suggesting different mechanisms for the amelioration of joint destruction and synovitis by E2.
Collapse
|
23
|
Spies CM, Wiebe E, Tu J, Li A, Gaber T, Huscher D, Seibel MJ, Zhou H, Buttgereit F. Acute murine antigen-induced arthritis is not affected by disruption of osteoblastic glucocorticoid signalling. BMC Musculoskelet Disord 2014; 15:31. [PMID: 24491163 PMCID: PMC3922092 DOI: 10.1186/1471-2474-15-31] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Accepted: 01/29/2014] [Indexed: 01/04/2023] Open
Abstract
Background The role of endogenous glucocorticoids (GC) in the initiation and maintenance of rheumatoid arthritis (RA) remains unclear. We demonstrated previously that disruption of GC signalling in osteoblasts results in a profound attenuation of K/BxN serum-induced arthritis, a mouse model of RA. To determine whether or not the modulation of the inflammatory response by osteoblasts involves T cells, we studied the effects of disrupted osteoblastic GC-signalling in the T cell-dependent model of antigen-induced arthritis (AIA). Methods Acute arthritis was induced in pre-immunised 11-week-old male 11β-hydroxysteroid dehydrogenase type 2 transgenic (tg) mice and their wild-type (WT) littermates by intra-articular injection of methylated bovine serum albumine (mBSA) into one knee joint. Knee diameter was measured every 1–2 days until euthanasia on day 14 post injection. In a separate experiment, arthritis was maintained for 28 days by weekly reinjections of mBSA. Tissues were analysed by histology, histomorphometry and microfocal-computed tomography. Serum cytokines levels were determined by multiplex suspension array. Results In both short and long term experiments, arthritis developed in tg and WT mice with no significant difference between both groups. Histological indices of inflammation, cartilage damage and bone erosion were similar in tg and WT mice. Bone volume and turnover at the contralateral tibia and systemic cytokine levels were not different. Conclusions Acute murine AIA is not affected by a disruption in osteoblastic GC signalling. These data indicate that osteoblasts do not modulate the T cell-mediated inflammatory response via a GC-dependent pathway.
Collapse
Affiliation(s)
- Cornelia M Spies
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Charitéplatz 1, D-10117 Berlin, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Chalise J, Narendra S, Paudyal B, Magnusson M. Interferon alpha inhibits antigen-specific production of proinflammatory cytokines and enhances antigen-specific transforming growth factor beta production in antigen-induced arthritis. Arthritis Res Ther 2013; 15:R143. [PMID: 24286140 PMCID: PMC3978460 DOI: 10.1186/ar4326] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Accepted: 09/17/2013] [Indexed: 02/06/2023] Open
Abstract
Introduction Interferon alpha (IFN-α) has a complex role in autoimmunity, in that it may both enhance and prevent inflammation. We have previously shown that the presence of IFN-α at sensitization protects against subsequent antigen-triggered arthritis. To understand this tolerogenic mechanism, we performed a descriptive, hypothesis-generating study of cellular and humoral responses associated with IFN-α-mediated protection against arthritis. Methods Arthritis was evaluated at day 28 in mice given a subcutaneous injection of methylated bovine serum albumin (mBSA), together with Freund adjuvant and 0 to 5,000 U IFN-α at days 1 and 7, followed by intraarticular injection of mBSA alone at day 21. The effect of IFN-α on mBSA-specific IgG1, IgG2a, IgG2b, IgA, and IgE was evaluated by enzyme-linked immunosorbent assay (ELISA). Cytokines in circulation and in ex vivo cultures on mBSA restimulation was evaluated with ELISA and Luminex, and the identity of cytokine-producing cells by fluorescence-activated cell sorting (FACS) analysis. Results Administration of IFN-α protected mice from arthritis in a dose-dependent manner but had no effect on antigen-specific antibody levels. However, IFN-α did inhibit the initial increase of IL-6, IL-10, IL-12, and TNF, and the recall response induced by intraarticular mBSA challenge of IL-1β, IL-10, IL-12, TNF, IFN-γ, and IL-17 in serum. IFN-α decreased both macrophage and CD4+ T cell-derived IFN-γ production, whereas IL-17 was decreased only in CD4+ T cells. Ex vivo, in mBSA-restimulated spleen and lymph node cell cultures, the inhibitory effect of in vivo administration of IFN-α on proinflammatory cytokine production was clearly apparent, but had a time limit. An earlier macrophage-derived, and stronger activation of the antiinflammatory cytokine transforming growth factor beta (TGF-β) was observed in IFN-α-treated animals, combined with an increase in CD4+ T cells producing TGF-β when arthritis was triggered by mBSA (day 21). Presence of IFN-α at immunizations also prevented the reduction in TGF-β production, which was induced by the intraarticular mBSA injection triggering arthritis in control animals. Conclusions Administration of IFN-α has a profound effect on the cellular response to mBSA plus adjuvant, but does not affect antigen-specific Ig production. By including IFN-α at immunizations, spleen and lymph node cells inhibit their repertoire of antigen-induced proinflammatory cytokines while enhancing antiinflammatory TGF-β production, first in macrophages, and later also in CD4+ T cells. On intraarticular antigen challenge, this antiinflammatory state is reenforced, manifested as inhibition of proinflammatory recall responses and preservation of TGF-β levels. This may explain why IFN-α protects against antigen-induced arthritis.
Collapse
|
25
|
Svensson MND, Andersson SEM, Erlandsson MC, Jonsson IM, Ekwall AKH, Andersson KME, Nilsson A, Bian L, Brisslert M, Bokarewa MI. Fms-like tyrosine kinase 3 ligand controls formation of regulatory T cells in autoimmune arthritis. PLoS One 2013; 8:e54884. [PMID: 23349985 PMCID: PMC3549988 DOI: 10.1371/journal.pone.0054884] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Accepted: 12/17/2012] [Indexed: 12/01/2022] Open
Abstract
Fms-like tyrosine kinase 3 ligand (Flt3L) is known as the primary differentiation and survival factor for dendritic cells (DCs). Furthermore, Flt3L is involved in the homeostatic feedback loop between DCs and regulatory T cell (Treg). We have previously shown that Flt3L accumulates in the synovial fluid in rheumatoid arthritis (RA) and that local exposure to Flt3L aggravates arthritis in mice, suggesting a possible involvement in RA pathogenesis. In the present study we investigated the role of Flt3L on DC populations, Tregs as well as inflammatory responses in experimental antigen-induced arthritis. Arthritis was induced in mBSA-immunized mice by local knee injection of mBSA and Flt3L was provided by daily intraperitoneal injections. Flow cytometry analysis of spleen and lymph nodes revealed an increased formation of DCs and subsequently Tregs in mice treated with Flt3L. Flt3L-treatment was also associated with a reduced production of mBSA specific antibodies and reduced levels of the pro-inflammatory cytokines IL-6 and TNF-α. Morphological evaluation of mBSA injected joints revealed reduced joint destruction in Flt3L treated mice. The role of DCs in mBSA arthritis was further challenged in an adoptive transfer experiment. Transfer of DCs in combination with T-cells from mBSA immunized mice, predisposed naïve recipients for arthritis and production of mBSA specific antibodies. We provide experimental evidence that Flt3L has potent immunoregulatory properties. Flt3L facilitates formation of Treg cells and by this mechanism reduces severity of antigen-induced arthritis in mice. We suggest that high systemic levels of Flt3L have potential to modulate autoreactivity and autoimmunity.
Collapse
Affiliation(s)
- Mattias N D Svensson
- Department of Rheumatology and Inflammation Research at Sahlgrenska Academy, University of Gothenburg, Göteborg, Sweden.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Peters MA, Wendholt D, Strietholt S, Frank S, Pundt N, Korb-Pap A, Joosten LAB, van den Berg WB, Kollias G, Eckes B, Pap T. The loss of α2β1 integrin suppresses joint inflammation and cartilage destruction in mouse models of rheumatoid arthritis. ACTA ACUST UNITED AC 2012; 64:1359-68. [PMID: 22083543 DOI: 10.1002/art.33487] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Integrin α2β1 functions as a major receptor for type I collagen on different cell types, including fibroblasts and inflammatory cells. Although in vitro data suggest a role for α2β1 integrin in regulating both cell attachment and expression of matrix-degrading enzymes such as matrix metalloproteinases (MMPs), mice that lack the α2 integrin subunit (Itga2(-/-) mice) develop normally and are fertile. We undertook this study to investigate the effect of Itga2 deficiency in 2 different mouse models of destructive arthritis: the antigen-induced arthritis (AIA) mouse model and the human tumor necrosis factor α (TNFα)-transgenic mouse model. METHODS AIA was induced in the knee joints of Itga2(-/-) mice and wild-type controls. Human TNF-transgenic mice were crossed with Itga2(-/-) mice and were assessed clinically and histopathologically for signs of arthritis, inflammation, bone erosion, and cartilage damage. MMP expression, proliferation, fibroblast attachment, and ERK activation were determined. RESULTS Under arthritic conditions, Itga2 deficiency led to decreased severity of joint pathology. Specifically, Itga2(-/-) mice showed less severe clinical symptoms and dramatically reduced pannus formation and cartilage erosion. Mice lacking α2β1 integrin exhibited reduced MMP-3 expression, both in their sera and in fibroblast-like synoviocytes (FLS), due to impaired ERK activation. Further, both the proliferation and attachment of FLS to cartilage were partially dependent on α2β1 integrin in vitro and in vivo. CONCLUSION Our findings suggest that α2β1 integrin contributes significantly to inflammatory cartilage destruction by promoting fibroblast proliferation and attachment and MMP expression.
Collapse
|
27
|
Ying F, Chalise JP, Narendra SC, Magnusson M. Type I IFN protects against antigen-induced arthritis. Eur J Immunol 2011; 41:1687-95. [PMID: 21469099 DOI: 10.1002/eji.201040956] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2010] [Revised: 02/17/2011] [Accepted: 03/22/2011] [Indexed: 12/22/2022]
Abstract
Autoimmune diseases including rheumatoid arthritis (RA) involve immune reactions against specific antigens. The type I IFN system is suspected to promote autoimmunity in systemic lupus erythematosus, but may also dampen immune reactions in e.g. inflammatory bowel disease. This prompted us to investigate the role of type I IFN in antigen-induced arthritis (AIA). The importance of type I IFN in methylated (m) BSA-induced arthritis was studied by using mice deficient for the type I IFN receptor (IFNAR) and by administration of the IFN-α activator viral double-stranded (ds) RNA or recombinant IFN-α at antigen sensitization. In IFNAR knock-out mice, arthritis severity was significantly higher than in WT mice. Administration of dsRNA at antigen sensitization protected WT but not IFNAR KO mice from arthritis. Also, addition of recombinant IFN-α during the immunization, but not the induction phase of arthritis, almost abolished arthritis. Protection mediated by IFN-α was accompanied by delayed and decreased antigen-specific proliferative responses, including impaired lymph node recall responses after intra-articular antigenic challenge. In conclusion, we demonstrate that type I IFN can prevent joint inflammation by downregulating antigen-specific cellular immunity.
Collapse
Affiliation(s)
- Fei Ying
- Affiliated Hospital of Guiyang Medical College, Department of Microbiology and Immunology, Guiyang, Guizhou, P R China
| | | | | | | |
Collapse
|
28
|
Teige A, Bockermann R, Hasan M, Olofsson KE, Liu Y, Issazadeh-Navikas S. CD1d-Dependent NKT Cells Play a Protective Role in Acute and Chronic Arthritis Models by Ameliorating Antigen-Specific Th1 Responses. THE JOURNAL OF IMMUNOLOGY 2010; 185:345-56. [DOI: 10.4049/jimmunol.0901693] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
29
|
Page CE, Smale S, Carty SM, Amos N, Lauder SN, Goodfellow RM, Richards PJ, Jones SA, Topley N, Williams AS. Interferon-gamma inhibits interleukin-1beta-induced matrix metalloproteinase production by synovial fibroblasts and protects articular cartilage in early arthritis. Arthritis Res Ther 2010; 12:R49. [PMID: 20307272 PMCID: PMC2888198 DOI: 10.1186/ar2960] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2009] [Revised: 02/18/2010] [Accepted: 03/22/2010] [Indexed: 02/07/2023] Open
Abstract
Introduction The first few months after symptom onset represents a pathologically distinct phase in rheumatoid arthritis (RA). We used relevant experimental models to define the pathological role of interferon-γ (IFN-γ) during early inflammatory arthritis. Methods We studied IFN-γ's capacity to modulate interleukin-1β (IL-1β) induced degenerative responses using RA fibroblast-like synoviocytes (FLS), a bovine articular cartilage explant (BACE)/RA-FLS co-culture model and an experimental inflammatory arthritis model (murine antigen-induced arthritis (AIA)). Results IFN-γ modulated IL-1β driven matrix metalloproteinases (MMP) synthesis resulting in the down-regulation of MMP-1 and MMP-3 production in vitro. IFN-γ did not affect IL-1β induced tissue inhibitor of metalloproteinase-1 (TIMP-1) production by RA FLS but skewed the MMP/TIMP-1 balance sufficiently to attenuate glycosaminoglycan-depletion in our BACE model. IFN-γ reduced IL-1β expression in the arthritic joint and prevented cartilage degeneration on Day 3 of AIA. Conclusions Early therapeutic intervention with IFN-γ may be critical to orchestrate tissue-protective responses during inflammatory arthritis.
Collapse
Affiliation(s)
- Charlotte E Page
- Section of Rheumatology, University Hospital of Wales, Cardiff and Vale NHS Trust, Cardiff, Wales, CF14 4XW, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Kolly L, Karababa M, Joosten LAB, Narayan S, Salvi R, Pétrilli V, Tschopp J, van den Berg WB, So AKL, Busso N. Inflammatory role of ASC in antigen-induced arthritis is independent of caspase-1, NALP-3, and IPAF. THE JOURNAL OF IMMUNOLOGY 2009; 183:4003-12. [PMID: 19717512 DOI: 10.4049/jimmunol.0802173] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Because IL-1beta plays an important role in inflammation in human and murine arthritis, we investigated the contribution of the inflammasome components ASC, NALP-3, IPAF, and caspase-1 to inflammatory arthritis. We first studied the phenotype of ASC-deficient and wild-type mice during Ag-induced arthritis (AIA). ASC(-/-) mice showed reduced severity of AIA, decreased levels of synovial IL-1beta, and diminished serum amyloid A levels. In contrast, mice deficient in NALP-3, IPAF, or caspase-1 did not show any alteration of joint inflammation, thus indicating that ASC associated effects on AIA are independent of the classical NALP-3 or IPAF inflammasomes. Because ASC is a ubiquitous cytoplasmic protein that has been implicated in multiple cellular processes, we explored other pathways through which ASC may modulate inflammation. Ag-specific proliferation of lymph node and spleen cells from ASC-deficient mice was significantly decreased in vitro, as was the production of IFN-gamma, whereas IL-10 production was enhanced. TCR ligation by anti-CD3 Abs in the presence or absence of anti-CD28 Abs induced a reduction in T cell proliferation in ASC(-/-) T cells compared with wild-type ones. In vivo lymph node cell proliferation was also significantly decreased in ASC(-/-) mice, but no effects on apoptosis were observed either in vitro or in vivo in these mice. In conclusion, these results strongly suggest that ASC modulates joint inflammation in AIA through its effects on cell-mediated immune responses but not via its implication in inflammasome formation.
Collapse
Affiliation(s)
- Laeticia Kolly
- Service of Rheumatology, Département de l'Appareil Locomoteur, Centre Hospitalier Universitaire Vaudois and University of Lausanne, University of Lausanne, Lausanne, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Bruns L, Frey O, Morawietz L, Landgraf C, Volkmer R, Kamradt T. Immunization with an immunodominant self-peptide derived from glucose-6-phosphate isomerase induces arthritis in DBA/1 mice. Arthritis Res Ther 2009; 11:R117. [PMID: 19640302 PMCID: PMC2745800 DOI: 10.1186/ar2777] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2008] [Revised: 06/23/2009] [Accepted: 07/29/2009] [Indexed: 12/19/2022] Open
Abstract
Introduction T-helper (Th) lymphocytes are critically required for the pathogenesis of glucose-6-phosphate isomerase (G6PI)-induced arthritis, but neither the G6PI epitopes recognized by arthritogenic T cells nor their pathogenic effector functions have been fully elucidated to date. We aimed at identifying arthritogenic G6PI peptides. Methods We used a library of overlapping peptides spanning the entire G6PI sequence to identify the epitopes recognized by G6PI-specific Th cells. Immunodominant peptides were then used to immunize mice. Arthritis development was evaluated clinically and histologically. The humoral and cellular immune responses upon peptide immunization were analyzed by ELISA and multiparameter flow cytometry, respectively. Results We identified six immunodominant T-cell epitopes in DBA/1 mice, of which three are arthritogenic. One of these peptides (G6PI469–483) is identical in man and mice. Immunization with this peptide induces arthritis, which is less severe and of shorter duration than arthritis induced by immunization with full-length G6PI. Upon immunization with either G6PI or peptide, the antigen-specific Th cells produce IL-17, RANKL, IFNγ and TNFα. Conclusions We identified immunodominant and arthritogenic epitopes of G6PI. Not all immunodominant peptides are arthritogenic. This is the first description of arthritis induced by immunization with a self-peptide in mice.
Collapse
Affiliation(s)
- Lisa Bruns
- Universitätsklinikum Jena, Institut für Immunologie, Leutragraben 3, Jena, Germany.
| | | | | | | | | | | |
Collapse
|
32
|
van Lent PLEM, Grevers LC, Blom AB, Arntz OJ, van de Loo FAJ, van der Kraan P, Abdollahi-Roodsaz S, Srikrishna G, Freeze H, Sloetjes A, Nacken W, Vogl T, Roth J, van den Berg WB. Stimulation of chondrocyte-mediated cartilage destruction by S100A8 in experimental murine arthritis. ACTA ACUST UNITED AC 2009; 58:3776-87. [PMID: 19035520 DOI: 10.1002/art.24074] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
OBJECTIVE To investigate whether S100A8 is actively involved in matrix metalloproteinase (MMP)-mediated chondrocyte activation. METHODS S100A8 and S100A9 proteins were detected in inflamed knee joints from mice with various forms of murine arthritis, using immunolocalization. Murine chondrocyte cell line H4 was stimulated with proinflammatory cytokines or recombinant S100A8. Messenger RNA (mRNA) and protein levels were measured using reverse transcriptase-polymerase chain reaction and intracellular fluorescence-activated cell sorting (FACS). Breakdown of aggrecan on the pericellular surface of the chondrocytes was measured using VDIPEN and NITEGE antibodies and FACS, and breakdown in patellar cartilage was measured by immunolocalization. RESULTS S100A8 and S100A9 proteins were abundantly expressed in and around chondrocytes in inflamed knee joints after induction of antigen-induced arthritis or onset of spontaneous arthritis in interleukin-1 (IL-1) receptor antagonist-knockout mice. Stimulation of chondrocytes by the proinflammatory cytokines tumor necrosis factor alpha, IL-1beta, IL-17, and interferon-gamma caused strong up-regulation of S100A8 mRNA and protein levels and up-regulation to a lesser extent of S100A9 levels. Stimulation of chondrocytes with S100A8 induced significant up-regulation of MMP-2, MMP-3, MMP-9, MMP-13, ADAMTS-4, and ADAMTS-5 mRNA levels (up-regulated 4, 4, 3, 16, 8, and 4 times, respectively). VDIPEN and NITEGE neoepitopes were significantly elevated in a concentration-dependent manner in chondrocytes treated with 0.2, 1, or 5 microg/ml of S100A8. (VDIPEN levels were elevated 17%, 67%, and 108%, respectively, and NITEGE levels were elevated 8%, 33%, and 67%, respectively.) S100A8 significantly increased the effect of IL-1beta on MMP-3, MMP-13, and ADAMTS-5. Mouse patellae incubated with both IL-1beta and S100A8 had elevated levels of NITEGE within the cartilage matrix when compared with patellae incubated with IL-1beta or S100A8 alone. CONCLUSION These findings indicate that S100A8 and S100A9 are found in and around chondrocytes in experimental arthritis. S100A8 up-regulates and activates MMPs and aggrecanase-mediated pericellular matrix degradation.
Collapse
Affiliation(s)
- P L E M van Lent
- Department of Rheumatology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|