1
|
Postlethwaite AE, Jiao Y, Yang C, Dong W, Aelion JA, Wang B, Postlethwaite BE, Sigal L, Kang AH, Myers LK, Wheller P, Ingels J, Gu W. Optimizing oral immune tolerance to Type II collagen in patients with rheumatoid arthritis: The importance of dose, interfering medication and genetics. Am J Med Sci 2024; 368:300-310. [PMID: 38897565 DOI: 10.1016/j.amjms.2024.06.008] [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: 01/19/2024] [Revised: 05/17/2024] [Accepted: 06/12/2024] [Indexed: 06/21/2024]
Abstract
OBJECTIVES Oral immune tolerance (OT) is a complex process with unknown genetic regulation. Our aim is to explore possible genetic control of OT in patients with rheumatoid arthritis (RA). METHODS RA patients with increased interferon γ production invitro when their isolated peripheral blood mononuclear cells (PBMC) were cultured with type II bovine collagen α1 chain [α1 (II)] were enrolled in this study and were randomly assigned to the "Low dose" type II collagen (CII) group (30 µg/day for 10 weeks, followed by 50 µg/day for 10 weeks, followed by 70 µg/day for 10 weeks) or "High dose" CII group (90 µg/day for 10 weeks, followed by 110 µg/day for 10 weeks, followed by 130 µg/day for 10 weeks). Heparinized blood was obtained at baseline and after each of the 10 weeks treatment for analysis of the invitro production of IFNγ by their PBMC stimulated by α1(II) . Single nucleotide polymorphism (SNP) analysis of the responders and non-responders to oral CII was conducted using GeneChip Mapping 10 K 2.0 Array. RESULTS The SNP A-15,737 was found to associate with the ability of CII to suppress IFNγ production by α1(CII)-stimulated RA PBMC. The potential for SNP A-15,737 to associate with the OT response for patients with another autoimmune disease [OT induced by oral type I bovine collagen (CI) in patients with diffuse cutaneous systemic sclersodid (dsSSc)] was also explored. CONCLUSIONS The ROT1 region plays a role in the control of IFNγ production after oral dosing of auto-antigens, thereby determining if oral tolerance to that antigen will develop.
Collapse
Affiliation(s)
- Arnold E Postlethwaite
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA; Department of Veterans Affairs Medical Center, Memphis, Tennessee, 38104, USA.
| | - Yan Jiao
- Departments of Orthopaedic Surgery and BME-Campbell Clinic, and Pathology, University of Tennessee Health Science Center (UTHSC), Memphis, TN 38163, USA
| | - Chengyuan Yang
- Departments of Orthopaedic Surgery and BME-Campbell Clinic, and Pathology, University of Tennessee Health Science Center (UTHSC), Memphis, TN 38163, USA
| | - Wei Dong
- Departments of Orthopaedic Surgery and BME-Campbell Clinic, and Pathology, University of Tennessee Health Science Center (UTHSC), Memphis, TN 38163, USA
| | - Jacob A Aelion
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Benjamin Wang
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | | | - Leonard Sigal
- Gateway Immunosciences, LLC, Stockbridge MA. 01262, USA
| | - Andrew H Kang
- Department of Veterans Affairs Medical Center, Memphis, Tennessee, 38104, USA
| | - Linda K Myers
- Department of Pediatrics University of Tennessee Health Science Center, Memphis, TN, USA
| | - Patricia Wheller
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Jesse Ingels
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Weikuan Gu
- Departments of Orthopaedic Surgery and BME-Campbell Clinic, and Pathology, University of Tennessee Health Science Center (UTHSC), Memphis, TN 38163, USA; Department of Veterans Affairs Medical Center, Memphis, Tennessee, 38104, USA
| |
Collapse
|
2
|
Radbruch A, Melchers F. [Why the regeneration of immunological tolerance by vaccination is difficult]. Z Rheumatol 2024; 83:105-111. [PMID: 38110746 DOI: 10.1007/s00393-023-01453-z] [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] [Accepted: 10/16/2023] [Indexed: 12/20/2023]
Abstract
Autoimmunity, including that involved in chronic inflammatory rheumatic diseases, seems to be the price we have to pay for our efficient immune system. It has the ability to precisely recognize pathogens and tumor cells, to efficiently fight them, to adapt to their alterations and provide specific immunity for a lifetime. "Inoculation", and more specifically "vaccination" takes advantage of this, either by transfer of protective antibodies (passive vaccination) or by using attenuated pathogens or parts of them by which a specific protective immunity is induced (active vaccination). The idea to use vaccination to reduce undesired (auto)immunity and chronic inflammation is nothing new in rheumatology. Many biologicals are antibodies, which specifically block the mediators of inflammation and in the broader sense are similar to a passive vaccination. The active vaccination with autoantigens using the recent mRNA/liposome technology, has shown in experimental animal models that they can prevent the formation of chronic inflammatory immune reactions, in that they strengthen the physiological tolerance and deviate the immune system to noninflammatory immune reactions against the antigen; however, there is still a long way to go to achieve the actual goals of a permanent suppression of established undesired immune reactions and the regeneration of immunological tolerance.
Collapse
Affiliation(s)
- Andreas Radbruch
- Deutsches Rheumaforschungszentrum Berlin, ein Leibniz Institut, Charitéplatz 1, 10117, Berlin, Deutschland.
| | - Fritz Melchers
- Deutsches Rheumaforschungszentrum Berlin, ein Leibniz Institut, Charitéplatz 1, 10117, Berlin, Deutschland
| |
Collapse
|
3
|
Ackun-Farmmer MA, Jewell CM. Delivery route considerations for designing antigen-specific biomaterial strategies to combat autoimmunity. ADVANCED NANOBIOMED RESEARCH 2023; 3:2200135. [PMID: 36938103 PMCID: PMC10019031 DOI: 10.1002/anbr.202200135] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Disease modifying drugs and biologics used to treat autoimmune diseases, although promising, are non-curative. As the field moves towards development of new approaches to treat autoimmune disease, antigen-specific therapies immunotherapies (ASITs) have emerged. Despite clinical approval of ASITs for allergies, clinical trials using soluble ASITs for autoimmunity have been largely unsuccessful. A major effort to address this shortcoming is the use of biomaterials to harness the features unique to specific delivery routes. This review focuses on biomaterials being developed for delivery route-specific strategies to induce antigen-specific responses in autoimmune diseases such as multiple sclerosis, type 1 diabetes, rheumatoid arthritis, and celiac disease. We first discuss the delivery strategies used in ongoing and completed clinical trials in autoimmune ASITs. Next, we highlight pre-clinical biomaterial approaches from the most recent 3 years in the context of these same delivery route considerations. Lastly, we provide discussion on the gaps remaining in biomaterials development and comment on the need to consider delivery routes in the process of designing biomaterials for ASITs.
Collapse
Affiliation(s)
- Marian A Ackun-Farmmer
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, USA
| | - Christopher M Jewell
- Fischell Department of Bioengineering, University of Maryland, College Park, MD, 20742, USA
- US Department of Veterans Affairs, VA Maryland Health Care System, Baltimore, MD, 21201, USA
- Robert E. Fischell Institute for Biomedical Devices, College Park, MD, 20742, USA
- Department of Microbiology and Immunology, University of Maryland Medical School, Baltimore, MD, 21201, USA
- Marlene and Stewart Greenebaum Cancer Center, Baltimore, MD, 21201, USA
| |
Collapse
|
4
|
Jabbari M, Barati M, Khodaei M, Babashahi M, Kalhori A, Tahmassian AH, Mosharkesh E, Arzhang P, Eini-Zinab H. Is collagen supplementation friend or foe in rheumatoid arthritis and osteoarthritis? A comprehensive systematic review. Int J Rheum Dis 2022; 25:973-981. [PMID: 35791039 DOI: 10.1111/1756-185x.14382] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/28/2022] [Accepted: 06/20/2022] [Indexed: 11/30/2022]
Abstract
Rheumatoid arthritis (RA) and osteoarthritis (OA) both are chronic diseases affecting joints. Immune response against collagen in both diseases may have a role in the initiation and progression of the disease. There is a hypothesis that suppression of immune response vs collagen could be a therapeutic approach in RA and OA. Exposure of gut immune system to collagen is a way to suppress immune response against collagen in the joints. So, the current systematic review is aimed to evaluate the effects of collagen supplementation in OA and RA patients. In the current systematic review, online electronic databases including PubMed/MEDLINE, Web of Sciences and Scopus were searched and finally 19 articles were included. The enrolled articles evaluated the effects of collagen supplementation on treatment of OA (n = 9) and RA (n = 10). Intact (n = 4) and hydrolyzed (n = 5) collagen were used to treat OA. All of the studies on RA used intact and type II collagen in their intervention. The last trials on collagen supplementation in RA and OA patients were performed in 2011 and 2016, respectively. High adverse effects of collagen supplementation and its low efficiency compared to routine treatments were reported by several included studies. Also, risk of bias assessment showed that most of the studies had poor quality. Therefore, it is not possible to definitely decide on the beneficial or detrimental effects of collagen supplementation on OA and RA patients. Further studies are needed to reach a final decision.
Collapse
Affiliation(s)
- Masoumeh Jabbari
- Student Research Committee, Department of Community Nutrition, National Nutrition and Food Technology Research Institute, Faculty of Nutrition and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Meisam Barati
- Department of Clinical Nutrition and Dietetics, National Nutrition and Food Technology Research Institute, Faculty of Nutrition and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Khodaei
- Student Research Committee, Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mina Babashahi
- Department of Community Nutrition, School of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Kalhori
- Nutrition Department, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Amir Hossein Tahmassian
- Department of Clinical Nutrition, Faculty of Nutrition, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Erfan Mosharkesh
- Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Pishva Arzhang
- Qods Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Hassan Eini-Zinab
- Department of Community Nutrition, National Nutrition and Food Technology Research Institute, Faculty of Nutrition Sciences and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Pinheiro-Rosa N, Torres L, Oliveira MDA, Andrade-Oliveira MF, Guimarães MADF, Coelho MM, Alves JDL, Maioli TU, Faria AMC. Oral tolerance as antigen-specific immunotherapy. IMMUNOTHERAPY ADVANCES 2021; 1:ltab017. [PMID: 35919733 PMCID: PMC9327124 DOI: 10.1093/immadv/ltab017] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 07/23/2021] [Accepted: 08/23/2021] [Indexed: 12/19/2022] Open
Abstract
Summary
Oral tolerance is a physiological phenomenon described more than a century ago as a suppressive immune response to antigens that gain access to the body by the oral route. It is a robust and long-lasting event with local and systemic effects in which the generation of mucosally induced regulatory T cells (iTreg) plays an essential role. The idea of using oral tolerance to inhibit autoimmune and allergic diseases by oral administration of target antigens was an important development that was successfully tested in 1980s. Since then, several studies have shown that feeding specific antigens can be used to prevent and control chronic inflammatory diseases in both animal models and clinically. Therefore, oral tolerance can be classified as an antigen-specific form of oral immunotherapy (OIT). In the light of novel findings on mechanisms, sites of induction and factors affecting oral tolerance, this review will focus on specific characteristics of oral tolerance induction and how they impact in its therapeutic application.
Collapse
Affiliation(s)
- Natália Pinheiro-Rosa
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Lícia Torres
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Mariana de Almeida Oliveira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Marcos Felipe Andrade-Oliveira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Mauro Andrade de Freitas Guimarães
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Monique Macedo Coelho
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Juliana de Lima Alves
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Tatiani Uceli Maioli
- Departamento de Nutrição, Escola de Enfermagem, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ana M Caetano Faria
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| |
Collapse
|
6
|
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: 13] [Impact Index Per Article: 4.3] [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
|
7
|
Norin U, Rintisch C, Meng L, Forster F, Ekman D, Tuncel J, Klocke K, Bäcklund J, Yang M, Bonner MY, Lahore GF, James J, Shchetynsky K, Bergquist M, Gjertsson I, Hubner N, Bäckdahl L, Holmdahl R. Endophilin A2 deficiency protects rodents from autoimmune arthritis by modulating T cell activation. Nat Commun 2021; 12:610. [PMID: 33504785 PMCID: PMC7840939 DOI: 10.1038/s41467-020-20586-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 12/07/2020] [Indexed: 11/18/2022] Open
Abstract
The introduction of the CTLA-4 recombinant fusion protein has demonstrated therapeutic effects by selectively modulating T-cell activation in rheumatoid arthritis. Here we show, using a forward genetic approach, that a mutation in the SH3gl1 gene encoding the endocytic protein Endophilin A2 is associated with the development of arthritis in rodents. Defective expression of SH3gl1 affects T cell effector functions and alters the activation threshold of autoreactive T cells, thereby leading to complete protection from chronic autoimmune inflammatory disease in both mice and rats. We further show that SH3GL1 regulates human T cell signaling and T cell receptor internalization, and its expression is upregulated in rheumatoid arthritis patients. Collectively our data identify SH3GL1 as a key regulator of T cell activation, and as a potential target for treatment of autoimmune diseases. The autoimmune disorder, rheumatoid arthritis (RA), has been associated with multiple pathophysiological factors. Here the authors show that deficiency in endophilin A2 in rodents protects them from experimental arthritis by altering T cell activation threshold and effector functions, thereby hinting a potential target for RA therapy.
Collapse
Affiliation(s)
- Ulrika Norin
- Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden.
| | - Carola Rintisch
- Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden.,Medical Inflammation Research, Lund University, Lund, Sweden.,Cardiovascular and Metabolic Sciences, Max-Delbrück-Center for Molecular Medicine (MDC), Berlin, Germany
| | - Liesu Meng
- Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden.,The Second affiliated hospital to Xi'an Jiaotong University and the Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 710061, Xi'an, Shaanxi, China
| | - Florian Forster
- Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Diana Ekman
- Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden.,Department of Biochemistry and Biophysics, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Stockholm University, Stockholm, Sweden
| | - Jonatan Tuncel
- Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Katrin Klocke
- Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Johan Bäcklund
- Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Min Yang
- Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Michael Y Bonner
- Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Gonzalo Fernandez Lahore
- Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Jaime James
- Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Klementy Shchetynsky
- Rheumatology Unit, Department of Medicine, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| | - Maria Bergquist
- Department of Rheumatology and Inflammation Research, Institute for Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Medical Sciences, Clinical Physiology, Uppsala University, Uppsala, Sweden
| | - Inger Gjertsson
- Department of Rheumatology and Inflammation Research, Institute for Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Norbert Hubner
- Cardiovascular and Metabolic Sciences, Max-Delbrück-Center for Molecular Medicine (MDC), Berlin, Germany.,Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Liselotte Bäckdahl
- Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Rikard Holmdahl
- Medical Inflammation Research, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden. .,The Second affiliated hospital to Xi'an Jiaotong University and the Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, 710061, Xi'an, Shaanxi, China.
| |
Collapse
|
8
|
Gencoglu H, Orhan C, Sahin E, Sahin K. Undenatured Type II Collagen (UC-II) in Joint Health and Disease: A Review on the Current Knowledge of Companion Animals. Animals (Basel) 2020; 10:E697. [PMID: 32316397 PMCID: PMC7222752 DOI: 10.3390/ani10040697] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/04/2020] [Accepted: 04/06/2020] [Indexed: 12/14/2022] Open
Abstract
OA is quite common in companion animals, especially in large breed dogs and horses. Collagen, the most abundant protein of mammals, has specific connective tissue types for skin, bones, reticulate, basal lamina, bones, cell surfaces, while type II collagen (UC-II) forms the main structure of cartilage tissue. Even at the smaller dosages, UC-II has also been reported to be more effective than the glucosamine and chondroitin sulfate supplements, which are the supplements most frequently used in the market. In this review, we summarize the effects of UC-II on joint health and function in health and disease conditions in companion animals.
Collapse
Affiliation(s)
- Hasan Gencoglu
- Department of Biology, Faculty of Science, Firat University, Elazig (+90) 424, Turkey;
| | - Cemal Orhan
- Department of Animal Nutrition, Faculty of Veterinary Medicine, Firat University, Elazig (+90) 424, Turkey; (C.O.); (E.S.)
| | - Emre Sahin
- Department of Animal Nutrition, Faculty of Veterinary Medicine, Firat University, Elazig (+90) 424, Turkey; (C.O.); (E.S.)
| | - Kazim Sahin
- Department of Animal Nutrition, Faculty of Veterinary Medicine, Firat University, Elazig (+90) 424, Turkey; (C.O.); (E.S.)
| |
Collapse
|
9
|
Yoon TW, Kim YI, Cho H, Brand DD, Rosloniec EF, Myers LK, Postlethwaite AE, Hasty KA, Stuart JM, Yi AK. Ameliorating effects of Gö6976, a pharmacological agent that inhibits protein kinase D, on collagen-induced arthritis. PLoS One 2019; 14:e0226145. [PMID: 31809526 PMCID: PMC6897462 DOI: 10.1371/journal.pone.0226145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 11/20/2019] [Indexed: 12/16/2022] Open
Abstract
Toll-like receptor (TLR) signaling can contribute to the pathogenesis of arthritis. Disruption of TLR signaling at early stages of arthritis might thereby provide an opportunity to halt the disease progression and ameliorate outcomes. We previously found that Gö6976 inhibits TLR-mediated cytokine production in human and mouse macrophages by inhibiting TLR-dependent activation of protein kinase D1 (PKD1), and that PKD1 is essential for proinflammatory responses mediated by MyD88-dependent TLRs. In this study, we investigated whether PKD1 contributes to TLR-mediated proinflammatory responses in human synovial cells, and whether Gö6976 treatment can suppress the development and progression of type II collagen (CII)-induced arthritis (CIA) in mouse. We found that TLR/IL-1R ligands induced activation of PKD1 in human fibroblast-like synoviocytes (HFLS). TLR/IL-1R-induced expression of cytokines/chemokines was substantially inhibited in Gö6976-treated HFLS and PKD1-knockdown HFLS. In addition, serum levels of anti-CII IgG antibodies, and the incidence and severity of arthritis after CII immunization were significantly reduced in mice treated daily with Gö6976. Synergistic effects of T-cell receptor and TLR, as well as TLR alone, on spleen cell proliferation and cytokine production were significantly inhibited in the presence of Gö6976. Our results suggest a possibility that ameliorating effects of Gö6976 on CIA may be due to its ability to inhibit TLR/IL-1R-activated PKD1, which might play an important role in proinflammatory responses in arthritis, and that PKD1 could be a therapeutic target for inflammatory arthritis.
Collapse
Affiliation(s)
- Tae Won Yoon
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Young-In Kim
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Hongsik Cho
- Department of Orthopedic Surgery and Biomedical Engineering, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - David D. Brand
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- Veterans Affairs Medical Center-Memphis, Memphis, Tennessee, United States of America
| | - Edward F. Rosloniec
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- Veterans Affairs Medical Center-Memphis, Memphis, Tennessee, United States of America
| | - Linda K. Myers
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Arnold E. Postlethwaite
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- Veterans Affairs Medical Center-Memphis, Memphis, Tennessee, United States of America
| | - Karen A. Hasty
- Department of Orthopedic Surgery and Biomedical Engineering, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- Veterans Affairs Medical Center-Memphis, Memphis, Tennessee, United States of America
| | - John M. Stuart
- Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- Veterans Affairs Medical Center-Memphis, Memphis, Tennessee, United States of America
| | - Ae-Kyung Yi
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- * E-mail:
| |
Collapse
|
10
|
Sieper J, Braun J. [Lessons learnt from 15 years of investigator-initiated trials on axial spondyloarthritis]. Z Rheumatol 2015; 74:125-31. [PMID: 25805515 DOI: 10.1007/s00393-014-1452-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
BACKGROUND Clinical research is receiving an increasing amount of attention and is essential for improving treatment of patients with rheumatic diseases. AIMS This article reports on 15 years of experience with conducting investigator-initiated studies on axial spondyloarthritis including ankylosing spondylitis. RESULTS We have organized and successfully conducted a series of open-labelled and placebo-controlled double-blind treatment studies and also non-interventional studies on this topic. The installation of a qualified and motivated trial unit and intensive collaboration with a statistician were crucial requirements for success. These results have increased our knowledge about the disease and changed and improved the diagnostic possibilities and the therapeutic options. CONCLUSION Investigator-initiated trials are an important link between basic and clinical research and can substantially contribute to improvement of patient care. This kind of research should be more systematically funded in the future.
Collapse
Affiliation(s)
- J Sieper
- Med. Klinik I, Rheumatologie, Charité Universitätsmedizin Berlin, Campus Benjamin Franklin, Hindenburgdamm 30, 12200, Berlin, Deutschland,
| | | |
Collapse
|
11
|
|
12
|
Thomas R. Dendritic cells and the promise of antigen-specific therapy in rheumatoid arthritis. Arthritis Res Ther 2013; 15:204. [PMID: 23374912 PMCID: PMC3672739 DOI: 10.1186/ar4130] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Rheumatoid arthritis (RA) is a systemic inflammatory disease resulting from an autoimmune response to self-antigens, leading to inflammation of synovial tissue of joints and subsequent cartilage and bone erosion. Current disease-modifying anti-rheumatic drugs and biologic inhibitors of TNF, IL-6, T cells and B cells block inflammation nonspecifically, which may lead to adverse effects, including infection. They do not generally induce long-term drug-free remission or restoration of immune tolerance to self-antigens, and lifelong treatment is usual. The development of antigen-specific strategies in RA has so far been limited by insufficient knowledge of autoantigens, of the autoimmune pathogenesis of RA and of the mechanisms of immune tolerance in man. Effective tolerance-inducing antigen-specific immunotherapeutic strategies hold promise of greater specificity, of lower toxicity and of a longer-term solution for controlling or even preventing RA. This paper reviews current understanding of autoantigens and their relationship to immunopathogenesis of RA, and emerging therapeutics that aim to leverage normal tolerance mechanisms for implementation of antigen-specific therapy in RA.
Collapse
|
13
|
Durrani K, Zakka FR, Ahmed M, Memon M, Siddique SS, Foster CS. Systemic Therapy With Conventional and Novel Immunomodulatory Agents for Ocular Inflammatory Disease. Surv Ophthalmol 2011; 56:474-510. [DOI: 10.1016/j.survophthal.2011.05.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2010] [Revised: 05/18/2011] [Accepted: 05/24/2011] [Indexed: 12/19/2022]
|
14
|
Macfarlane GJ, El-Metwally A, De Silva V, Ernst E, Dowds GL, Moots RJ. Evidence for the efficacy of complementary and alternative medicines in the management of rheumatoid arthritis: a systematic review. Rheumatology (Oxford) 2011; 50:1672-83. [PMID: 21652584 DOI: 10.1093/rheumatology/ker119] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE To critically evaluate the evidence regarding complementary and alternative medicine (CAM) taken orally or applied topically (excluding fish oil) in the treatment of RA. METHODS Randomized controlled trials (RCTs) of RA using CAMs, in comparison with other treatments or placebo, published in English up to August 2010, were eligible for inclusion. They were identified using systematic searches of bibliographic databases and manual searching of reference lists. Information was extracted on outcomes and statistical significance, in comparison with alternative treatments, and reported side effects. The methodological quality of the primary studies was determined using the Jadad scoring system. RESULTS Reported RCTs were available for 18 CAMs in the management of RA. There was no consistent evidence available for any of the reviewed substances to suggest that they were efficacious as complementary medicines to standard treatment. Nevertheless, the studies conducted on borage seed oil (n = 2) and thunder god vine (n = 3) have been positive and may warrant further investigation. Not all CAM compounds studied were free of major adverse effects. CONCLUSION The major limitation in reviewing the evidence for CAMs is the paucity of RCTs in the area. The available evidence does not support their current use in the management of RA.
Collapse
Affiliation(s)
- Gary J Macfarlane
- Epidemiology Group, School of Medicine and Dentistry, University of Aberdeen, Polwarth Building (Room 1:071), Foresterhill, Aberdeen, Scotland AB25 2ZD, UK.
| | | | | | | | | | | |
Collapse
|
15
|
|
16
|
Immunotherapies. Rheumatology (Oxford) 2011. [DOI: 10.1016/b978-0-323-06551-1.00057-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
17
|
|
18
|
Xinqiang S, Fei L, Nan L, Yuan L, Fang Y, Hong X, Lixin T, Juan L, Xiao Z, Yuying S, Yongzhi X. Therapeutic efficacy of experimental rheumatoid arthritis with low-dose methotrexate by increasing partially CD4+CD25+Treg cells and inducing Th1 to Th2 shift in both cells and cytokines. Biomed Pharmacother 2010; 64:463-71. [DOI: 10.1016/j.biopha.2010.01.007] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2009] [Accepted: 01/25/2010] [Indexed: 02/06/2023] Open
|
19
|
Wehrens EJ, van Wijk F, Roord ST, Albani S, Prakken BJ. Treating arthritis by immunomodulation: is there a role for regulatory T cells? Rheumatology (Oxford) 2010; 49:1632-44. [PMID: 20463189 DOI: 10.1093/rheumatology/keq130] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The discovery of regulatory T cells almost 15 years ago initiated a new and exciting research area. The growing evidence for a critical role of these cells in controlling autoimmune responses has raised expectations for therapeutic application of regulatory T cells in patients with autoimmune arthritis. Here, we review recent studies investigating the presence, phenotype and function of these cells in patients with RA and juvenile idiopathic arthritis (JIA) and consider their therapeutic potential. Both direct and indirect methods to target these cells will be discussed. Arguably, a therapeutic approach that combines multiple regulatory T-cell-enhancing strategies could be most successful for clinical application.
Collapse
Affiliation(s)
- Ellen J Wehrens
- Department of Pediatric Immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht (UMCU), Lundlaan 6, 3584 EA, Utrecht, The Netherlands.
| | | | | | | | | |
Collapse
|
20
|
Serological investigation of IgG levels and subclasses in rheumatoid arthritis patients following ingestion of bovine type II collagen: results of a double blind, randomised controlled trial. Clin Rheumatol 2010; 30:193-9. [PMID: 20440528 DOI: 10.1007/s10067-010-1473-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2009] [Revised: 01/19/2010] [Accepted: 04/19/2010] [Indexed: 10/19/2022]
Abstract
The purpose of this study was to investigate whether clinical improvement in patient with rheumatoid arthritis (RA) taking oral Bovine type II collagen (bCII) is associated with changes in levels of anti-bCII total IgG antibody and its subclasses. Four groups were given either 5, 0.5 or 0.05 mg of bCII or a placebo in a double-blind randomised control trial. The sera IgG anti-collagen type II and antibodies of the IgG subclasses, IgG1-4, were measured at the start and end of the trial by ELISA. Total IgG anti-collagen II antibodies in the pooled active treatment groups were statistically significantly reduced compared with the placebo group (p = 0.021). Decreasing total IgG titres were observed in the 0.5-mg group (p = 0.008), 0.05-mg group (p = 0.004) and 5-mg group (p = 0.078) before and after treatment. For IgG1, IgG2, IgG3 and IgG4 subclasses, in the 0.5-mg group in which the best clinical response was observed, there was statistically significant decreases observed in the IgG2 and IgG3 subclasses (p = 0.047, p = 0.046). Treatment with bCII in patients with RA led to a reduction in anti-collagen II antibody titre indicating an active biological effect as observed previously in animal model of RA. The largest decrease in total and subclasses of anti-collagen antibody titres occurred in the groups of patients with the best therapeutic response to bCII, supporting the conclusion of the clinical trial and suggests that immune regulation explains the therapeutic effect.
Collapse
|
21
|
Immunotherapy of Rheumatoid Arthritis Targeting Inflammatory Cytokines and Autoreactive T Cells. Arch Immunol Ther Exp (Warsz) 2010; 58:27-36. [DOI: 10.1007/s00005-009-0058-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2009] [Accepted: 07/06/2009] [Indexed: 12/19/2022]
|
22
|
Wei W, Zhang LL, Xu JH, Xiao F, Bao CD, Ni LQ, Li XF, Wu YQ, Sun LY, Zhang RH, Sun BL, Xu SQ, Liu S, Zhang W, Shen J, Liu HX, Wang RC. A multicenter, double-blind, randomized, controlled phase III clinical trial of chicken type II collagen in rheumatoid arthritis. Arthritis Res Ther 2009; 11:R180. [PMID: 19951408 PMCID: PMC3003530 DOI: 10.1186/ar2870] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2009] [Revised: 09/29/2009] [Accepted: 12/01/2009] [Indexed: 11/19/2022] Open
Abstract
Introduction Chicken type II collagen (CCII) is a protein extracted from the cartilage of chicken breast and exhibits intriguing possibilities for the treatment of autoimmune diseases by inducing oral tolerance. A 24-week, double-blind, double-dummy, randomized, methotrexate (MTX)-controlled study was conducted to evaluate the efficacy and safety of CCII in the treatment of rheumatoid arthritis (RA). Methods Five hundred three RA patients were included in the study. Patients received either 0.1 mg daily of CCII (n = 326) or 10 mg once a week of MTX (n = 177) for 24 weeks. Each patient was evaluated for pain, morning stiffness, tender joint count, swollen joint count, health assessment questionnaire (HAQ), assessments by investigator and patient, erythrocyte sedimentation rate (ESR), and C-reactive protein (CRP) by using the standard tools at baseline (week 0) and at weeks 12 and 24. Additionally, rheumatoid factor (RF) was evaluated at weeks 0 and 24. Measurement of a battery of biochemical parameters in serum, hematological parameters, and urine analysis was performed to evaluate the safety of CCII. Results Four hundred fifty-four patients (94.43%) completed the 24-week follow-up. In both groups, there were decreases in pain, morning stiffness, tender joint count, swollen joint count, HAQ, and assessments by investigator and patient, and all differences were statistically significant. In the MTX group, ESR and CRP decreased. RF did not change in either group. At 24 weeks, 41.55% of patients in the CCII group and 57.86% in the MTX group met the American College of Rheumatology 20% improvement criteria (ACR-20) and 16.89% and 30.82%, respectively, met the ACR 50% improvement criteria (ACR-50). Both response rates for ACR-20 and ACR-50 in the CCII group were lower than those of the MTX group, and this difference was statistically significant (P < 0.05). The DAS28 (disease activity score using 28 joint counts) values of the two treatment groups were calculated, and there was a statistically significant difference between the two treatment groups (P < 0.05). Gastrointestinal complaints were common in both groups, but there were fewer and milder side effects in the CCII group than in the MTX group. The incidence of adverse events between the two groups was statistically significant (P < 0.05). Conclusions CCII is effective in the treatment of RA and is safe for human consumption. CCII exerts its beneficial effects by controlling inflammatory responses through inducing oral tolerance in RA patients. Trials Registration Clinical trial registration number: ChiCTR-TRC-00000093.
Collapse
Affiliation(s)
- Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immunopharmacology of Education Ministry, 81 Meishan Road, Hefei 230032, PR China.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
GUPTA RC, CANERDY TD, SKAGGS P, STOCKER A, ZYRKOWSKI G, BURKE R, WEGFORD K, GOAD JT, ROHDE K, BARNETT D, DeWEES W, BAGCHI M, BAGCHI D. Therapeutic efficacy of undenatured type-II collagen (UC-II) in comparison to glucosamine and chondroitin in arthritic horses. J Vet Pharmacol Ther 2009; 32:577-84. [DOI: 10.1111/j.1365-2885.2009.01079.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
24
|
Type II collagen oral tolerance; mechanism and role in collagen-induced arthritis and rheumatoid arthritis. Mod Rheumatol 2009; 19:581-9. [PMID: 19697097 DOI: 10.1007/s10165-009-0210-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2009] [Accepted: 07/08/2009] [Indexed: 12/24/2022]
Abstract
Oral tolerance means a diminished immune response to previously fed antigens. Repeated oral administrations of type II collagen (CII) induce oral tolerance and inhibit the development of collagen-induced arthritis (CIA). Dendritic cells (DCs) in the gut-associated lymphoid tissue (GALT) take up the CII and then present it to T cells to generate regulatory T cells (Tregs), which induce systemic immune tolerance to CII. Inhibitory cytokines, such as transforming growth factor (TGF)-beta and interleukin (IL)-10, and several immune regulatory molecules, including indoleamine 2,3-dioxygenase (IDO) and retinoic acid, play an important role in Treg generation. Each DC subset may play different roles, and CD11c+CD11b+DCs and IDO+DCs are important in the generation of antigen-inducible Tregs in CII oral tolerance. Upon stimulation with the antigen involved in its generation, Treg is activated and regulates the immune response through inhibitory cytokine production, cell-to-cell contact-dependent mechanisms, DC modification, and bystander suppression. The DCs and Tregs are deeply involved in oral tolerance through reciprocal interactions. Several clinical trials have been conducted in RA patients to examine the efficacy of CII oral tolerance. An understanding the mechanism of oral tolerance to CII would give clinicians new insights into the development of natural immune tolerance and new therapeutic approaches for the treatment of autoimmune diseases.
Collapse
|
25
|
Toussirot E, Robinet E, Saas P, Chabod J, Augé B, Cozma G, Tiberghien P, Roudier J, Wendling D. Bacterial extract (OM-89) specific and non specific immunomodulation in rheumatoid arthritis patients. Autoimmunity 2009; 39:299-306. [PMID: 16891218 DOI: 10.1080/08916930600738425] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
The Escherichia Coli bacterial extract (OM-89) is used in the treatment of rheumatoid arthritis (RA). We evaluated the immunological changes induced by oral administration of OM-89 in 12 RA patients (polyclonal T cell reactivity to PHA, T cell precursor frequencies specific for OM-89 and Tetanus toxoid (TT), a control antigen and the release of Th1 (IFN-gamma, TNF-alpha), Th2 (IL-4) and T regulatory 1 cell (Tr1) (IL-10) cytokines in the supernatants of PBMC cultures. Stimulation index in response to PHA decreased at month 3 as well as T cell precursor frequencies specific for TT with similar trends for OM-89-specific T cell precursor frequencies. OM-89 induced a strong production of IL-10, a significant decrease in IL-4 production while TNF-alpha and IFN-gamma production tended to decrease during the study. Our results suggest that OM-89 has immunomodulatory properties by inducing changes in PBMC cytokines release suggestive of an induced Tr1 response to OM-89.
Collapse
Affiliation(s)
- Eric Toussirot
- Department of Rheumatology, University Hospital Jean Minjoz, Bd Fleming, F-25030 Besançon cédex, France.
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Hasselberg A, Schön K, Tarkowski A, Lycke N. Role of CTA1R7K-COL-DD as a novel therapeutic mucosal tolerance-inducing vector for treatment of collagen-induced arthritis. ACTA ACUST UNITED AC 2009; 60:1672-82. [DOI: 10.1002/art.24566] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
27
|
Song X, Liang F, Liu N, Luo Y, Xue H, Yuan F, Tan L, Sun Y, Xi C, Xi Y. Construction and characterization of a novel DNA vaccine that is potent antigen-specific tolerizing therapy for experimental arthritis by increasing CD4+CD25+Treg cells and inducing Th1 to Th2 shift in both cells and cytokines. Vaccine 2008; 27:690-700. [PMID: 19095031 DOI: 10.1016/j.vaccine.2008.11.090] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2008] [Revised: 10/31/2008] [Accepted: 11/12/2008] [Indexed: 01/04/2023]
Abstract
Currently available treatments for rheumatoid arthritis (RA) are often ineffective in ameliorating the progression of disease, particularly the invasive destruction of articular cartilage and bone, and RA remains incurable. Therefore, vaccinotherapy of RA with an antigen-specific tolerizing DNA vaccine may offer new promise for overcoming this difficulty. Using recombinant technology, the DNA sequences encoding chicken type II collagen (CCOL2A1) with deleted N-propeptides were obtained from the plasmid pPIC9K/pCalpha(1)(II), and then cloned into pcDNA3.1(+). The resulting recombinant plasmid pcDNA-CCOL2A1 was produced in Escherichia coli, purified, characterized and used as a tolerizing DNA vaccine for the treatment of collagen-induced arthritis (CIA). Therapeutic efficacy and potential action mechanisms of pcDNA-CCOL2A1 tolerizing DNA vaccine against CIA were studied. Here we demonstrate that a single intravenous treatment with novel tolerizing DNA vaccine pcDNA-CCOL2A1 can induce potent immune tolerance against CIA. The efficacy of this therapy was verified by clinical visual scoring, radiographic X-ray, histopathological examination, and anti-CII IgG levels. Furthermore, the action mechanism behind this efficacy can be at least partially attributed to increased CD4(+)CD25(+) T regulatory cells, which specifically down-modulate the T lymphocyte proliferative response to CCII, induce a shift of Th1 to Th2 cells, as well as down-regulate Th1-cytokine TNF-alpha, while up-regulating both Th2-cytokine IL-10 and Th3-cytokine TGF-beta. More importantly, pcDNA-CCOL2A1 alone seems to be as effective as the current "golden standard" treatment, methotrexate (MTX). Taken together, these results suggest that we have successfully developed a novel tolerizing DNA vaccine encoding CCII, which is the first description of a tolerizing DNA vaccine encoding CCII for antigen-specific tolerizing therapy but not prophylactic against CIA.
Collapse
Affiliation(s)
- Xinqiang Song
- Department of Immunology and National Center for Biomedicine Analysis, Beijing 307 Hospital Affiliated to Academy of Medical Sciences, No. 8 Dongda Ave., Fengtai District, Beijing 100071, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Xi C, Tan L, Sun Y, Liang F, Liu N, Xue H, Luo Y, Yuan F, Sun Y, Xi Y. A novel recombinant peptide containing only two T-cell tolerance epitopes of chicken type II collagen that suppresses collagen-induced arthritis. Mol Immunol 2008; 46:729-37. [PMID: 19041137 DOI: 10.1016/j.molimm.2008.10.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2008] [Revised: 10/06/2008] [Accepted: 10/08/2008] [Indexed: 01/05/2023]
Abstract
Immunotherapy of rheumatoid arthritis (RA) using oral-dosed native chicken or bovine type II collagen (nCII) to induce specific immune tolerance is an attractive strategy. However, the majority of clinical trials of oral tolerance in human diseases including RA in recent years have been disappointing. Here, we describe a novel recombinant peptide rcCTE1-2 which contains only two tolerogenic epitopes (CTE1 and CTE2) of chicken type II collagen (cCII). These are the critical T-cell determinants for suppression of RA that were first developed and used to compare its suppressive effects with ncCII on the collagen-induced arthritis (CIA) model. The rcCTE1-2 was produced using the prokaryotic pET expression system and purified by Ni-NTA His affinity chromatography. Strikingly, our results showed clearly that rcCTE1-2 was as efficacious as ncCII at the dose of 50 microg/kg/d. This dose significantly reduced footpad swelling, arthritic incidence and scores, and deferred the onset of disease. Furthermore, rcCTE1-2 of 50 microg/kg/d could lower the level of anti-nCII antibody in the serum of CIA animals, decrease Th1-cytokine INF-gamma level, and increase Th3-cytokine TGF-beta(1) produced level by spleen cells from CIA mice after in vivo stimulation with ncCII. Importantly, rcCTE1-2 was even more potent than native cCII, which was used in the clinic for RA. Equally importantly, the findings that the major T-cell determinants of cCII that are also recognized by H-2(b) MHC-restricted T cells have not previously been reported. Taken together, these results suggest that we have successfully developed a novel recombinant peptide rcCTE1-2 that can induce a potent tolerogenic response in CIA.
Collapse
Affiliation(s)
- Caixia Xi
- Department of Immunology and National Center for Biomedicine Analysis, Beijing 307 Hospital Affiliated to Academy of Medical Sciences, No. 8, Dongda Ave, Fengtai District, Beijing 100071, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
D'Altilio M, Peal A, Alvey M, Simms C, Curtsinger A, Gupta RC, Canerdy TD, Goad JT, Bagchi M, Bagchi D. Therapeutic Efficacy and Safety of Undenatured Type II Collagen Singly or in Combination with Glucosamine and Chondroitin in Arthritic Dogs. Toxicol Mech Methods 2008; 17:189-96. [DOI: 10.1080/15376510600910469] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
30
|
So JS, Lee CG, Kwon HK, Yi HJ, Chae CS, Park JA, Hwang KC, Im SH. Lactobacillus casei potentiates induction of oral tolerance in experimental arthritis. Mol Immunol 2008; 46:172-80. [PMID: 18804867 DOI: 10.1016/j.molimm.2008.07.038] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2008] [Revised: 07/16/2008] [Accepted: 07/17/2008] [Indexed: 12/31/2022]
Abstract
Probiotics have been shown to exert beneficial effects on modulation of diverse diseases. However, no information is available for the effect of probiotics in the induction of oral tolerance in autoimmune diseases. The main purpose of this study was to elucidate whether Lactobacillus casei (L. casei) affect the induction of oral tolerance in experimental rheumatoid arthritis (RA). Type II collagen (CII) alone or together with L. casei was orally administered into collagen-induced arthritis (CIA) rats, and its effects on the clinical and histopathological aspects of RA were investigated. Co-administration of L. casei with CII more effectively suppressed clinical symptoms, paw swelling, lymphocyte infiltration and destruction of cartilage tissues of experimental arthritis than the rats treated with CII alone. The enhanced therapeutic efficacy was associated with an increase in anti-inflammatory cytokines (IL-10 and TGF-beta) while decreasing pro-inflammatory cytokines (IL-1beta, IL-2, IL-6, IL-12, IL-17, IFN-gamma and TNF-alpha). Co-administration of L. casei with CII more effectively suppressed CII-reactive T cell proliferation and the levels of Th1-type IgG isotypes (IgG2a and IgG2b), while up-regulating Foxp3 expression levels and the population of Foxp3(+) CD4(+) T cells. Our study provides evidence that L. casei could potentiate antigen-specific oral tolerance and suppress Th1-type immune responses of arthritic inflammation.
Collapse
Affiliation(s)
- Jae-Seon So
- Department of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju 500-712, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Postlethwaite AE, Wong WK, Clements P, Chatterjee S, Fessler BJ, Kang AH, Korn J, Mayes M, Merkel PA, Molitor JA, Moreland L, Rothfield N, Simms RW, Smith EA, Spiera R, Steen V, Warrington K, White B, Wigley F, Furst DE. A multicenter, randomized, double-blind, placebo-controlled trial of oral type I collagen treatment in patients with diffuse cutaneous systemic sclerosis: I. oral type I collagen does not improve skin in all patients, but may improve skin in late-phase disease. ACTA ACUST UNITED AC 2008; 58:1810-22. [PMID: 18512816 DOI: 10.1002/art.23501] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE To investigate the safety and efficacy of oral bovine type I collagen (CI) treatment in patients who have had diffuse cutaneous systemic sclerosis (dcSSc; scleroderma) for <or=10 years. METHODS One hundred sixty-eight patients with dcSSc were enrolled in a double-blind, placebo-controlled trial of oral CI (500 microg/day) or placebo administered over 12 months, with a followup visit at month 15. The primary outcome was the modified Rodnan skin thickness score (MRSS). Other clinical and immune system parameters were also assessed. RESULTS Intent-to-treat and modified intent-to-treat analyses showed that for the total population of patients with dcSSc, there were no significant differences in the mean change in MRSS or other key clinical parameters between the CI and placebo treatment groups at 12 months or at 15 months. However, in a subanalysis of the available data at month 15, the CI-treated group of patients with late-phase dcSSc experienced a significant reduction in the MRSS compared with that in the placebo-treated patients with late-phase dcSSc (change in MRSS at month 15 -7.9 versus -2.9; P = 0.0063). CONCLUSION Although the results from this trial did not meet the primary outcome goals, the findings from exploratory analyses indicated that CI treatment may benefit patients with late-phase dcSSc. This new treatment strategy and preliminary clinical observations in patients with dcSSc need to be corroborated.
Collapse
|
32
|
Abstract
INTRODUCTION Rheumatoid arthritis is a common inflammatory condition. A large number of patients seek alternative or complementary therapies of which diet is an important component. This article reviews the evidence for diet in rheumatoid arthritis along with the associated concept of oral tolerization. METHODS References were taken from Medline from 1966 to September 2004. The keywords, rheumatoid arthritis, diet, n-3 fatty acids, vitamins, and oral tolerization, were used. RESULTS Randomized controlled trials (RCTs) indicate that dietary supplementation with n-3 fatty acids provides modest symptomatic benefit in groups of patients with rheumatoid arthritis. Epidemiological studies and RCTs show cardiovascular benefits in the broader population and patients with ischemic heart disease. A number of mechanisms through which n-3 fats may reduce inflammation have been identified. In a small number of patients with rheumatoid arthritis, other dietary manipulation such as fasting, vegan, and elimination diets may have some benefit. However, many of these diets are impractical or difficult to sustain long term. CONCLUSIONS Dietary manipulation provides a means by which patients can a regain a sense of control over their disease. Dietary n-3 supplementation is practical and can be easily achieved with encapsulated or, less expensively, bottled fish oil.
Collapse
Affiliation(s)
- Lisa K Stamp
- Department of Medicine, Christchurch School of Medicine and Health Sciences, University of Otago, New Zealand.
| | | | | |
Collapse
|
33
|
Xi C, Liu N, Liang F, Guo S, Sun Y, Yang F, Xi Y. Molecular cloning, characterization and localization of chicken type II procollagen gene. Gene 2006; 366:67-76. [PMID: 16297573 DOI: 10.1016/j.gene.2005.06.032] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2004] [Revised: 04/13/2005] [Accepted: 06/02/2005] [Indexed: 11/21/2022]
Abstract
Chicken type II procollagen (ccol2a1) has become as an important oral tolerance protein for effective treatment of rheumatoid arthritis. However, its molecular identity remains unclear. Here, we reported the full-length cDNA and nearly complete genomic DNA encoding ccol2a1. We have determined the structural organization, evolutional characters, developmental expression and chromosomal mapping of the gene. The full-length cDNA sequence spans 4837 bp containing all the coding region of the ccol2a1 including 3' and 5' untranslation region. The deduced peptide of ccol2a1, composed of 1420 amino acids, can be divided into signal peptide, N-propeptide, N-telopeptide, triple helix, C-telopeptide and C-propeptide. The ccol2a1 genomic DNA sequence was determined to be 12,523 bp long containing 54 exons interrupted by 53 introns. Comparison of the ccol2a1 with its counterparts in human, mouse, canine, horse, rat, frog and newt revealed highly conserved sequence in the triple helix domain. Chromosomal mapping of ccol2a1 locates it on 4P2. While the ccol2a1 mRNA was expressed in multiple tissues, the protein was only detected in chondrogenic cartilage, vitreous body and cornea. The ccol2a1 was found to contain two isoforms detected by RT-PCR. The distribution of the ccol2a1 lacking exon 2wasfrequently detected in chondrogenic tissues, whereas the exon 2-containing isoform was more abundant in non-chondrogenic tissues. These results provide useful information for preparing recombinant chicken type II collagen and for a better understanding of normal cartilage development.
Collapse
Affiliation(s)
- Caixia Xi
- Department of Immunology and National Center for Biomedicine Analysis, Beijing 307 Hospital Affiliated to Academy of Medical Sciences, No. 8 Dong-Da Street, Beijing, 100071, PR China
| | | | | | | | | | | | | |
Collapse
|
34
|
Deparle LA, Gupta RC, Canerdy TD, Goad JT, D'Altilio M, Bagchi M, Bagchi D. Efficacy and safety of glycosylated undenatured type-II collagen (UC-II) in therapy of arthritic dogs. J Vet Pharmacol Ther 2005; 28:385-90. [PMID: 16050819 DOI: 10.1111/j.1365-2885.2005.00668.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
DeParle L. A., Gupta R. C., Canerdy T. D., Goad J. T., D'Altilio M., Bagchi M., Bagchi D. Efficacy and safety of glycosylated undenatured type-II collagen (UC-II) in therapy of arthritic dogs. J. vet. Pharmacol. Therap.28, 385-390. In large breed dogs, arthritis is very common because of obesity, injury, aging, immune disorder, or genetic predispositions. This study was therefore undertaken to evaluate clinical efficacy and safety of undenatured type-II collagen (UC-II) in obese-arthritic dogs. Fifteen dogs in three groups received either no UC-II (Group I) or UC-II with 1 mg/day (Group II) or 10 mg/day (Group III) for 90 days. Lameness and pain were measured on a weekly basis for 120 days (90 days treatment plus 30 days post-treatment). Blood samples were assayed for creatinine and blood urea nitrogen (markers of renal injury); and alanine aminotransferase and aspartate aminotransferase (evidence of hepatic injury). Dogs receiving 1 mg or 10 mg UC-II/day for 90 days showed significant declines in overall pain and pain during limb manipulation and lameness after physical exertion, with 10 mg showed greater improvement. At either dose of UC-II, no adverse effects were noted and no significant changes were noted in serum chemistry, suggesting that UC-II was well tolerated. In addition, dogs receiving UC-II for 90 days showed increased physical activity level. Following UC-II withdrawal for a period of 30 days, all dogs experienced a relapse of overall pain, exercise-associated lameness, and pain upon limb manipulation. These results suggest that daily treatment of arthritic dogs with UC-II ameliorates signs and symptoms of arthritis, and UC-II is well tolerated as no adverse effects were noted.
Collapse
Affiliation(s)
- L A Deparle
- Murray State University, Murray/Hopkinsville, KY 42241-2000, USA
| | | | | | | | | | | | | |
Collapse
|
35
|
Lee WK, Park JY, Jung S, Yang CW, Kim WU, Kim HY, Park JH, Park JS. Preparation and characterization of biodegradable nanoparticles entrapping immunodominant peptide conjugated with PEG for oral tolerance induction. J Control Release 2005; 105:77-88. [PMID: 15919128 DOI: 10.1016/j.jconrel.2005.03.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2004] [Revised: 03/09/2005] [Accepted: 03/18/2005] [Indexed: 11/20/2022]
Abstract
PEG-conjugated immunodominant peptides for collagen-induced arthritis (CIA) were prepared for oral tolerance induction instead of whole Type II collagen (CII), because a small peptide can be converted to a macromolecule soluble in methylene chloride by the coupling of poly-ethylene glycol (PEG). PEG-pep1 was synthesized from a peptide and mPEG-NH2 (Mw approximately 5000) using SPDP as a linker, whereas PEG-pep2 was prepared by the direct disulfide coupling between PEG-OD (Mw approximately 10,000) and the peptide. PEG-pep1 and PEG-pep2 were purified by gel permeation chromatography (GPC), and the peak fractions of GPC were identified by GPC and MALDI-TOF mass spectroscopy. The peptide coupling gave much earlier retention times for PEG-pep1 (11.26 min) and PEG-pep2 (10.61 min) than for mPEG-SPDP (15.63 min) and mPEG-OD (14.58 min). The Mw's of mPEG-NH2, mPEG-SPDP, PEG-pep1, mPEG-OD and PEG-pep2 were 5451, 5588, 7035, 10,360 and 11,826, respectively, suggesting that PEG-pep1 and PEG-pep2 of high purity could be obtained. The nanoparticles entrapping PEG-pep1 and PEG-pep2 (NP/PEG-pep1 and NP/PEG-pep2) were prepared by the o/w solvent evaporation method, whereas the peptide-loaded nanoparticles (NP/pep) were prepared by the w/o/w double emulsion method. Although all the nanoparticles had a similar spherical morphology under scanning electron microscopy, NP/pep showed up as having a larger mean size than the others, which was confirmed by dynamic light scattering analysis (NP/pep, 499.7+/-27.2 nm; NP/PEG-pep1, 333.0+/-16.8 nm; NP/PEG-pep2, 342.4+/-15.1 nm). The lower encapsulation efficiency of NP/pep (21.0+/-1.6%) than NP/PEG-pep1 (66.5+/-5.0%) and NP/PEG-pep2 (73.8+/-5.5%) can also be attributed to the preparation method. In in vitro release studies, NP/PEG-pep1 and NP/PEG-pep2 displayed a similar release profile, close to a linear release pattern, whereas NP/pep displayed a tri-phasic release profile. From these results, it was demonstrated that nanoparticles entrapping a PEG-conjugated peptide could be an alternative delivery method for the induction of oral tolerance rather than CII and peptide.
Collapse
Affiliation(s)
- Woo-kyoung Lee
- School of Nano Engineering, Inje University, Kimhae, Korea
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Staines NA, Derry CJ, Marinova-Mutafchieva L, Ali N, Davies DH, Murphy JJ. Constraints on the efficacy of mucosal tolerance in treatment of human and animal arthritic diseases. Ann N Y Acad Sci 2005; 1029:250-9. [PMID: 15681763 DOI: 10.1196/annals.1309.056] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Mucosal administration of an autoantigen has been shown to be a powerful way of inducing tolerance in both animal and human arthritis clinical trials. Bovine or chicken type II collagen has been administered orally to rheumatoid arthritis patients, resulting in some, although in many cases rather limited, clinical improvement. Animal studies have revealed that the mechanisms that underlie induction of mucosal tolerance include clonal deletion, suppression of the proinflammatory Th1 cells, and the induction of regulatory T cells. These cells, defined as a persistently CD25-expressing subset of CD4(+) cells, are frequently anergic, may produce anti-inflammatory cytokines such as IL-10 and TGF-beta, and are likely to be agents of bystander suppression. A key feature that may affect the induction of these cells and other suppressive mechanisms is the dose of antigen administered. The results from human clinical trials suggest a daily dose of significantly less than 1 mg is optimal. Similarly data from collagen-induced arthritis studies reveal an optimal dose above and below which there is little or no immune suppression. Indeed, the incorrect dose can prime the immune response and aggravate disease. The timing and frequency of administration is also vital to the level of immune tolerance induced and the control of the pathological process. This and other findings derived from animal studies are discussed here in relation to the results from human clinical trials.
Collapse
Affiliation(s)
- Norman A Staines
- Infection and Immunity Research Group, King's College London, Stamford Street, London, SE1 9NN, UK.
| | | | | | | | | | | |
Collapse
|
37
|
|
38
|
Albani S, Prakken B. T cell epitope–specific immune therapy for rheumatic diseases. ACTA ACUST UNITED AC 2005; 54:19-25. [PMID: 16385493 DOI: 10.1002/art.21520] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Salvatore Albani
- Departments of Medicine and Pediatrics, University of California-San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA.
| | | |
Collapse
|
39
|
Abstract
Almost all current therapeutic concepts in autoimmune diseases are based on the systemic suppression of immune functions and are not curative. The recent introduction of biologicals such as tumour necrosis factor blocking antibodies or receptors has added greater specificity to efficient management of disease by targeted suppression of rheumatic inflammation. It is evident, however, that only the elimination of the cells secreting inflammatory mediators, rather than the blockade of secreted molecules, will offer real specific therapeutic advantages in the future. Merely the elimination of such cells and also cells controlling the secreting effector cells could be curative and induce true long term remissions. We review here the state of the art and future therapeutic concepts that are based on the specific modulation of pathogenic cells that induce and sustain autoimmune inflammation. This sounds visionary, however, a variety of basic tools are at hand now. Thus, direct and specific cell therapy of rheumatic diseases will become a true alternative to conventional therapies.
Collapse
Affiliation(s)
- A Radbruch
- German Rheumatism Research Centre Berlin, Schumannstr. 21/22, 10117 Berlin, Germany.
| | | |
Collapse
|
40
|
Kamradt T, Schubert D. The role and clinical implications of G6PI in experimental models of rheumatoid arthritis. Arthritis Res Ther 2004; 7:20-8. [PMID: 15642150 PMCID: PMC1064898 DOI: 10.1186/ar1476] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The antigens that trigger the pathogenic immune response in rheumatoid arthritis (RA) remain unknown. Until recently it was assumed that either viral or microbial antigens, or joint-specific antigens were the target of arthritogenic T and B lymphocytes in RA. Consequently, murine models of arthritis are induced by immunization with either joint-specific antigens such as type II collagen or microbial products such as streptococcal cell wall. In the K/B×N T-cell receptor transgenic mouse model arthritis is caused by a systemic autoimmune response to the ubiquitously expressed glycolytic enzyme glucose-6-phosphate isomerase (G6PI). The autoreactive transgenic T cells recognize G6PI and provide help for the production of arthritogenic IgG antibodies against G6PI. More recently it was shown that G6PI immunization induces severe symmetrical peripheral polyarthritis in genetically unaltered DBA/I mice. In that model CD4+ T cells are necessary not only for the induction but also for the effector phase of arthritis. Here we review the pathomechanisms that lead from systemic autoreactivity to arthritis in these models, consider the relevance of anti-G6PI immune reactivity for RA, and discuss the insights into the pathogenesis of RA and possibly other autoimmune conditions that can be gained from these models.
Collapse
MESH Headings
- Animals
- Antibody Specificity
- Arthritis, Experimental/enzymology
- Arthritis, Experimental/etiology
- Arthritis, Experimental/immunology
- Arthritis, Rheumatoid/immunology
- Autoantibodies/immunology
- Autoantigens/immunology
- Autoimmune Diseases/enzymology
- Autoimmune Diseases/etiology
- Autoimmune Diseases/immunology
- B-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/immunology
- Cell Wall/chemistry
- Cell Wall/immunology
- Collagen Type II/immunology
- Collagen Type II/toxicity
- Complement System Proteins/immunology
- Crosses, Genetic
- Glucose-6-Phosphate Isomerase/immunology
- Histocompatibility Antigens Class II/immunology
- Humans
- Immunization
- Immunization, Passive
- Immunoglobulin G/immunology
- Interleukin-1/physiology
- Mast Cells/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Inbred DBA
- Mice, Inbred NOD
- Mice, Transgenic
- Nervous System Autoimmune Disease, Experimental/etiology
- Nervous System Autoimmune Disease, Experimental/immunology
- Neutrophils/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
Collapse
Affiliation(s)
- Thomas Kamradt
- Institut für Immunologie, Klinikum der Friedrich-Schiller Universität Jena, Jena, Germany.
| | | |
Collapse
|
41
|
Svendsen P, Andersen CB, Willcox N, Coyle AJ, Holmdahl R, Kamradt T, Fugger L. Tracking of Proinflammatory Collagen-Specific T Cells in Early and Late Collagen-Induced Arthritis in Humanized Mice. THE JOURNAL OF IMMUNOLOGY 2004; 173:7037-45. [PMID: 15557201 DOI: 10.4049/jimmunol.173.11.7037] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Rheumatoid arthritis is a chronic inflammatory disease associated with certain HLA-DR4 subtypes. The target autoantigen(s) is unknown, but type II collagen (CII) is a candidate, with a single immunodominant DR4-restricted 261-273 T cell epitope (CII(261-273)). In the present study, we have prepared HLA-DR4:CII(261-273) tetramers and analyzed peripheral blood, lymph node, and synovial fluid cells from DR4-transgenic mice with early and late collagen-induced arthritis to draw a fuller picture of the role of CII-reactive Th cells in disease development. Their frequencies increased approximately 20-fold in blood 1-2 wk postimmunization, and even more in acutely arthritic joints. Our data strongly suggest that CII-specific Th cells are necessary, but not sufficient for collagen-induced arthritis. The CII-specific Th cells displayed an activated proinflammatory Th1 phenotype, and their expansion correlated with onset and severity of arthritis and also with anti-CII Ab levels. Surprisingly, shortly after the first clinical signs of arthritis, activated HLA-DR4:CII tetramer(+) cells became undetectable in the synovial fluid and rare in the blood, but persisted in lymph nodes. Consequently, future human studies should focus on patients with early arthritis, and on their synovial cells, to re-evaluate the occurrence and pathogenic importance of CII-specific or other Th cells in rheumatoid arthritis.
Collapse
MESH Headings
- Acute Disease
- Animals
- Arthritis, Experimental/genetics
- Arthritis, Experimental/immunology
- Arthritis, Experimental/pathology
- Chronic Disease
- Collagen Type II/administration & dosage
- Collagen Type II/immunology
- Disease Progression
- Epitopes, T-Lymphocyte/administration & dosage
- Epitopes, T-Lymphocyte/genetics
- Epitopes, T-Lymphocyte/immunology
- Genetic Vectors
- HLA-DR4 Antigen/administration & dosage
- HLA-DR4 Antigen/biosynthesis
- HLA-DR4 Antigen/genetics
- HLA-DR4 Antigen/immunology
- Humans
- Immunoglobulin G/biosynthesis
- Immunoglobulin G/blood
- Immunophenotyping
- Inflammation Mediators/administration & dosage
- Inflammation Mediators/immunology
- Leukocytes, Mononuclear/chemistry
- Lymphocyte Count
- Mice
- Mice, Inbred DBA
- Mice, Transgenic
- Peptide Fragments/administration & dosage
- Peptide Fragments/biosynthesis
- Peptide Fragments/genetics
- Peptide Fragments/immunology
- Severity of Illness Index
- Synovial Fluid/cytology
- Synovial Fluid/immunology
- Synovial Membrane/immunology
- Synovial Membrane/pathology
- T-Lymphocytes, Helper-Inducer/immunology
- T-Lymphocytes, Helper-Inducer/pathology
Collapse
Affiliation(s)
- Pia Svendsen
- Department of Clinical Immunology, Aarhus University Hospital, Skejby Sygehus, Aarhus, Denmark
| | | | | | | | | | | | | |
Collapse
|
42
|
Ganesan N, Chegu H, Chandrasekaran AN. Effect of type II collagen treatment on the antioxidant status in immune tissues of adjuvant induced arthritic rats. Indian J Clin Biochem 2003; 18:216-22. [PMID: 23105415 PMCID: PMC3453862 DOI: 10.1007/bf02867390] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Adjuvant induced arthritis (AIA) is a model widely used to study Rheumatoid arthritis (RA). In the present study, lipid peroxides level in spleen and thymus of AIA rats was observed to be significantly high compared to normal rats. A significant decrease in ascorbic acid (ASA), reduced glutathione (GSH), superoxide dismutase activity (SOD) was also observed in spleen and thymus of AIA rats compared to normal rats. There was also a steady increase in the circulating immune complex level (CIC) throughout the experimental period in serum of AIA rats. In the present investigation, it was decided to study the effect of pre and post treatment with TYPE II collagen on the antioxidant status and the circulating immune complex level in AIA rats. The results from the present work indicates that the pretreatment with TYPE II collagen was effective in bringing significant changes on all the parameters studied in AIA rats. The post treatment with TYPE II collagen was effective in bringing significant changes on the CIC immune complex level and GSH content in the thymus tissue of AIA rats. The present work suggests that the pre treatment with TYPE II collagen was more effective in suppressing the disease than the post treatment.
Collapse
Affiliation(s)
- Nalini Ganesan
- Dept. of Biochemistry, SRMC & RI (DU), Porur, 600 116 Chennai
| | | | | |
Collapse
|
43
|
Ding CH, Li Q, Xiong ZY, Zhou AW, Jones G, Xu SY. Oral administration of type II collagen suppresses pro-inflammatory mediator production by synoviocytes in rats with adjuvant arthritis. Clin Exp Immunol 2003; 132:416-23. [PMID: 12780687 PMCID: PMC1808728 DOI: 10.1046/j.1365-2249.2003.02167.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The objective of this study was to investigate the effect of the oral administration of type II collagen (CII) on pro-inflammatory mediator production by synoviocytes in rats with adjuvant arthritis (AA). Sprague-Dawley rats were fed with bovine CII either before immunization with Complete Freund's adjuvant (CFA) or after initiation of arthritis. Hind paw secondary swelling was measured and synoviocytes were harvested. Sera from portal vein of oral tolerized rats were collected and in vitro synoviocytes culture or synoviocytes-Peyer's Patches (PP) cells coculture system were developed. Interleukin (IL)-1 activity was measured by a mouse thymocyte activation assayed by MTT dye reduction and tumour necrosis factor (TNF) activity was measured by an L929 cytotoxicity bioassay. Nitric oxide (NO) and malondialdehyde (MDA) levels were measured by biochemical methods. We found that feeding with CII (5, 50 and 500 micro g/kg) for 7 days before immunization significantly suppressed hind paw secondary swelling measured at day 16, 20, 24 and 28 (all P < 0.01) and pro-inflammatory mediator (IL-1, TNF, NO and MDA) production by synoviocytes (all P < 0.01) in rats with AA. Feeding with CII (5, 50 and 500 micro g/kg) for 7 days after initiation of arthritis had a similar effect. CII (1, 10, 100 micro g/ml) had no effect on IL-1 and TNF production by synoviocytes in vitro, but CII 10 micro g/ml suppressed IL-1 and TNF production by synoviocytes-PP cells coculture system (P < 0.01), which was antagonized by anti-TGF-beta antibody (10 micro g/ml) (P < 0.01). Portal serum (1 : 10) from oral tolerized rats suppressed IL-1 and TNF production by synoviocytes (P < 0.01), which was also antagonized by anti-TGF-beta antibody (10 micro g/ml) (P < 0.01). We conclude that oral administration of CII had prophylactic and therapeutic effects on AA and over-production of IL-1, TNF, NO and MDA by synoviocytes was suppressed. Bystander active suppression may be the main mechanism of oral CII in the suppression of synoviocyte function.
Collapse
Affiliation(s)
- C H Ding
- Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China.
| | | | | | | | | | | |
Collapse
|
44
|
Thurau SR, Wildner G. An HLA-peptide mimics organ-specific antigen in autoimmune uveitis: its role in pathogenesis and therapeutic induction of oral tolerance. Autoimmun Rev 2003; 2:171-6. [PMID: 12848942 DOI: 10.1016/s1568-9972(03)00011-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Autoimmune uveitis is a sight threatening disease, which is conventionally treated with immunosuppressive medication. New treatment strategies include immunological approaches and aim at antigen specificity like oral tolerance. A peptide from the sequence of certain HLA-class I molecules plays a central role in the pathogenesis. When T cells recognize the HLA-peptide and are activated they are enabled to pass the blood-retina barrier. In the eye they recognize a cross-reactive organ-specific peptide and cause inflammation, which presents as uveitis. Here, we used the HLA-peptide as oral tolerogen to treat uveitis patients in an open study. All patients showed a positive therapeutic response and could reduce their long-lasting conventional immunosuppressive treatment. We did not observe any side effects. Moreover, side effects from conventional therapy could be reduced significantly.
Collapse
Affiliation(s)
- Stephan R Thurau
- Section of Immunobiology, Department of Ophthalmology, Ludwig-Maximilians-University, Mathildenstr. 8, 80336 Munich, Germany.
| | | |
Collapse
|
45
|
Asai K, Hachimura S, Kimura M, Toraya T, Yamashita M, Nakayama T, Kaminogawa S. T cell hyporesponsiveness induced by oral administration of ovalbumin is associated with impaired NFAT nuclear translocation and p27kip1 degradation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:4723-31. [PMID: 12391180 DOI: 10.4049/jimmunol.169.9.4723] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Oral tolerance is an important physiological component of the immune system whereby the organism avoids dangerous reactions such as hypersensitivity to ingested food proteins and other luminal Ags which may cause tissue damage and inflammation. In addition, it has been shown in animal models and in humans that oral tolerance can be applied to controlling undesired immune responses, including autoimmune diseases, allergies, and organ transplant rejections. However, the molecular mechanisms of oral tolerance have been poorly defined. In this study, we investigated the molecular basis underlying the hyporesponsiveness of orally tolerant CD4 T cells using a TCR transgenic mouse system in which oral tolerance was induced by long-term feeding with high dose Ag. We demonstrate that the hyporesponsive state of the CD4 T cells was maintained by a selective impairment in the TCR-induced calcium/NFAT signaling pathway and in the IL-2R-induced degradation of p27(kip1) and cell cycle progression. Thus, physiological mucosal tolerance is revealed to be associated with a unique type of T cell hyporesponsiveness which differs from previously described anergic T cells.
Collapse
MESH Headings
- Active Transport, Cell Nucleus/drug effects
- Active Transport, Cell Nucleus/genetics
- Active Transport, Cell Nucleus/immunology
- Adaptor Proteins, Signal Transducing
- Administration, Oral
- Amino Acid Sequence
- Animals
- Antibody Formation/genetics
- CD4-Positive T-Lymphocytes/drug effects
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- Calcium Signaling/drug effects
- Calcium Signaling/genetics
- Calcium Signaling/immunology
- Carrier Proteins/metabolism
- Cell Cycle/genetics
- Cell Cycle/immunology
- Cell Cycle Proteins/metabolism
- Clonal Anergy/drug effects
- Clonal Anergy/genetics
- Cyclin-Dependent Kinase Inhibitor p27
- Cyclin-Dependent Kinases/antagonists & inhibitors
- DNA-Binding Proteins/antagonists & inhibitors
- DNA-Binding Proteins/metabolism
- Dose-Response Relationship, Immunologic
- Immediate-Early Proteins/biosynthesis
- Interleukin-2/pharmacology
- Ionomycin/pharmacology
- Isoenzymes/antagonists & inhibitors
- Isoenzymes/metabolism
- MAP Kinase Kinase 4
- MAP Kinase Signaling System/genetics
- MAP Kinase Signaling System/immunology
- Membrane Proteins/metabolism
- Mice
- Mice, Inbred BALB C
- Mice, Knockout
- Mice, Transgenic
- Milk Proteins
- Mitogen-Activated Protein Kinase Kinases/physiology
- Mitogen-Activated Protein Kinases/physiology
- Molecular Sequence Data
- NFATC Transcription Factors
- Nuclear Proteins
- Ovalbumin/administration & dosage
- Ovalbumin/immunology
- Phospholipase C gamma
- Phosphoproteins/metabolism
- Phosphorylation
- Protein-Tyrosine Kinases/metabolism
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Interleukin-2/biosynthesis
- STAT5 Transcription Factor
- Spleen/cytology
- Spleen/immunology
- Spleen/metabolism
- Suppressor of Cytokine Signaling Proteins
- Trans-Activators/metabolism
- Transcription Factors/antagonists & inhibitors
- Transcription Factors/metabolism
- Tumor Suppressor Proteins/metabolism
- Type C Phospholipases/antagonists & inhibitors
- Type C Phospholipases/metabolism
- Tyrosine/metabolism
- ZAP-70 Protein-Tyrosine Kinase
Collapse
Affiliation(s)
- Kazumi Asai
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, University of Tokyo, Japan
| | | | | | | | | | | | | |
Collapse
|
46
|
Thurau SR, Wildner G. Oral tolerance for treating uveitis - new hope for an old immunological mechanism. Prog Retin Eye Res 2002; 21:577-89. [PMID: 12433378 DOI: 10.1016/s1350-9462(02)00039-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Oral tolerance induction has evolved as an attractive approach for the treatment of autoimmune uveitis. This approach is effective and generally void of the side effects associated with conventional immunosuppression. Following uptake of soluble antigen via the gut mucosa a specific systemic tolerance is generated. Experimental autoimmune diseases such as uveitis can efficiently be treated when autoantigens are fed to animals. The immunological mechanisms of oral tolerance are not well understood but are thought to involve the recognition of tolerogenic epitopes, generation of suppressor T cells and altered regulation of selected cytokines. The dose, purity of the antigen (tissue extract vs. single peptide) and concomitant treatment with cytokines were evaluated with the aim to enhance oral tolerance. Immunomodulatory drugs can abrogate oral tolerance. This requires careful evaluation with respect to therapeutic approaches in patients. The first clinical trials for treatment of uveitis with oral retinal autoantigen or an HLA-peptide crossreactive with S-Antigen show a promising therapeutic effect and confirmed the safety of this approach.
Collapse
Affiliation(s)
- Stephan R Thurau
- Department of Ophthalmology, Ludwig-Maximilians-University, Mathildenstr. 8, 80336, München, Germany
| | | |
Collapse
|
47
|
Furuzawa-Carballeda J, Rodríquez-Calderón R, Díaz de León L, Alcocer-Varela J. Mediators of inflammation are down-regulated while apoptosis is up-regulated in rheumatoid arthritis synovial tissue by polymerized collagen. Clin Exp Immunol 2002; 130:140-9. [PMID: 12296865 PMCID: PMC1906486 DOI: 10.1046/j.1365-2249.2002.01955.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2002] [Indexed: 11/20/2022] Open
Abstract
The aim of the study was to determine whether collagen-polyvinylpyrrolidone (collagen-PVP) modifies some proinflammatory responses in synovium cultures from rheumatoid arthritis (RA) patients. Synovium from 10 RA patients were cultured with or without 1% collagen-PVP. Tissues on the 3rd, 5th and 7th culture day were sectioned and stained by the Herovici technique. Total collagen and type I/III collagen ratios were evaluated by the Woessner micromethod and by interrupted gel electrophoresis, respectively. Collagenolytic activity was assessed by degradation of [3H]-collagen in supernatants. TIMP-1, IL-1beta and TNF-alpha were determined in supernatants by ELISA, and the results were normalized by DNA concentration. IL-1beta, TNF-alpha, IL-6, IL-8, MMP-1, TIMP-1, Cox-1, VCAM-1, ICAM-1 and Fas/APO95 expression was evaluated by immunohistochemistry. Apoptosis was detected by TUNEL technique. The histological analysis and electrophoresis revealed a 1.7-fold increase of type III collagen in a time-dependent fashion in collagen-PVP-treated cultures. Proinflammatory cytokines (IL-1beta: 58 +/- 9 versus 22 +/- 10; TNF-alpha: 41 +/- 6 versus 11 +/- 3; IL-8: 59 +/- 12 versus 29 +/- 9; treated versus untreated), adhesion molecule (ICAM-1: 57 +/- 11 versus 29 +/- 15; VCAM-1: 49 +/- 7 versus 21 +/- 13; treated versus untreated) as well as Cox-1 (59 +/- 10 versus 20 +/- 3) expression was down-regulated in RA synovium treated. Meanwhile, TIMP-1 (36 +/- 7 versus 57 +/- 11) and Fas expression (20 +/- 10 versus 55 +/- 13) and apoptosis (14 +/- 3 versus 55 +/- 5) were up-regulated in treated cultures compared with controls. In supernatants, the collagenolytic activity, as well as IL-1beta and TNF-alpha, levels were all down-regulated in treated cultures (two, three, fourfold, respectively). The addition of collagen-PVP to synovium-induced down-modulation of some inflammatory parameters and an increase in apoptosis of synovial cells. Perhaps this mechanism could contribute to inhibit outgrowth of pannus formation and to down-regulate inflammation of joints in patients with RA.
Collapse
Affiliation(s)
- J Furuzawa-Carballeda
- Department of Cellular Biology, Instituto de Investigaciones Biomédicas, UNAM, Mexico City, Mexico.
| | | | | | | |
Collapse
|
48
|
Kim WU, Lee WK, Ryoo JW, Kim SH, Kim J, Youn J, Min SY, Bae EY, Hwang SY, Park SH, Cho CS, Park JS, Kim HY. Suppression of collagen-induced arthritis by single administration of poly(lactic-co-glycolic acid) nanoparticles entrapping type II collagen: a novel treatment strategy for induction of oral tolerance. ARTHRITIS AND RHEUMATISM 2002; 46:1109-20. [PMID: 11953991 DOI: 10.1002/art.10198] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
OBJECTIVE Poly(lactic-co-glycolic acid) (PLGA), a biodegradable polymer, is a carrier for drug delivery systems. This study was undertaken to investigate the tolerogenic effect of single administration of PLGA entrapping type II collagen (CII) on the development of collagen-induced arthritis (CIA). METHODS The biophysical properties of PLGA nanoparticles entrapping CII (PLGA-CII) were investigated by in vitro release testing of CII, immunohistochemistry analysis, and electron microscopy. PLGA-CII was fed singly to animals 14 days before immunization, and the effect on joint inflammation was assessed. Circulating IgG anti-CII antibodies and T cell responses to CII in draining lymph nodes were assayed by enzyme-linked immunosorbent assay and (3)H-thymidine incorporation assay, respectively. The expression of messenger RNA (mRNA) for transforming growth factor beta (TGFbeta) and tumor necrosis factor alpha (TNFalpha) was determined by reverse transcriptase-polymerase chain reaction. RESULTS The in vitro release test showed that CII was slowly discharged from PLGA-CII over a period of a month. After single administration of PLGA-CII, numerous particles approximately 300 nm in size were detectable in Peyer's patches, by electron microscopy and immunohistochemical staining for CII, 14 days after the original feeding. Mice fed a single dose of PLGA containing 40 microg of CII had significantly reduced values for incidence and severity of arthritis, serum IgG anti-CII antibodies, and CII-specific T cell proliferation as compared with mice fed solvent alone, those fed 6 doses of 20 microg CII alone, and those fed a single dose of PLGA alone. PLGA-CII was also able to suppress CIA after disease onset. Moreover, PLGA-CII-fed mice showed a higher level of TGFbeta mRNA expression in Peyer's patches, but a lower level of TNFalpha mRNA expression in draining lymph nodes, compared with the other groups of mice. CONCLUSION Our data show that PLGA may serve as a powerful vehicle to promote the tolerance effect of oral CII and that single administration of PLGA-CII may hold promise as a new treatment strategy in rheumatoid arthritis.
Collapse
Affiliation(s)
- Wan-Uk Kim
- The Catholic University of Korea, Seoul, Republic of South Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Choy EH, Scott DL, Kingsley GH, Thomas S, Murphy AG, Staines N, Panayi GS. Control of rheumatoid arthritis by oral tolerance. ARTHRITIS AND RHEUMATISM 2001; 44:1993-7. [PMID: 11592359 DOI: 10.1002/1529-0131(200109)44:9<1993::aid-art347>3.0.co;2-a] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
OBJECTIVE Previous randomized controlled trials for treatment of rheumatoid arthritis (RA) with acid-soluble chicken and bovine type II collagen (CII) have produced conflicting results. This randomized, double-blind, controlled trial examined the therapeutic effect of bovine CII tablets in RA. METHODS CII tablets were prepared by adsorption onto a lactose base. Patients with a duration of RA of > or = 2 years and who had failed treatment with at least 1 slow-acting drug were recruited, provided that they had active arthritis. Patients were randomly assigned to receive either 0.05 mg, 0.5 mg, or 5 mg of CII or placebo daily for 6 months. All slow-acting drugs were stopped at least 4 weeks before starting CII, although prednisolone was permitted at dosages < 10 mg/day. Clinical assessments were performed at screening and at 0, 1, 4, 8, 12, 16, 20, and 24 weeks of treatment. RESULTS Fifty-five patients were recruited. Initially, there were no significant differences in mean Disease Activity Scores between groups. At 24 weeks, there was a significant difference (P = 0.041, by Kruskal-Wallis analysis of variance); the major components of this difference were attributable to relatively large decreases in the 0.5 mg CII group (19% of initial values) and to minimal decreases in patients receiving placebo (3% of initial values). Twenty patients had American College of Rheumatology 20% responses; 11 of these were in the 0.5 mg CII group and 3 were in each of the other groups, a significant difference (chi2 = 14.6, P = 0.002). There was no significant difference in any clinical measure between the placebo, 0.05 mg CII, and 5 mg CII groups. There were no side effects associated with CII treatment. CONCLUSION Treatment with 0.5 mg/day of bovine CII is well tolerated and produces small, but significant, disease improvement in RA. However, the therapeutic window is narrow. The difference between our results and those of other trials may relate to the dose, species, and formulation of the CII.
Collapse
Affiliation(s)
- E H Choy
- Guy's Kings, Hospital School of Medicine, King's College, London, UK
| | | | | | | | | | | | | |
Collapse
|
50
|
Derry CJ, Harper N, Davies DH, Murphy JJ, Staines NA. Importance of dose of type II collagen in suppression of collagen-induced arthritis by nasal tolerance. ARTHRITIS AND RHEUMATISM 2001; 44:1917-27. [PMID: 11508445 DOI: 10.1002/1529-0131(200108)44:8<1917::aid-art330>3.0.co;2-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
OBJECTIVE To determine the influence of the dose of collagen given nasally on the induction of specific mucosal tolerance in collagen-induced arthritis. METHODS The severity of clinical arthritis induced in DBA/1 mice was studied after the nasal administration (before disease induction) of 1 of 4 doses (across a 2-log range) of bovine type II collagen (CII). Parameters of immunity included lymphocyte proliferation and cytokine production in vitro in response to antigen stimulation, and the production of anticollagen IgG antibody subclasses. RESULTS The 3 highest doses (20, 80, and 320 microg) ameliorated disease severity, whereas the lowest dose (5 microg) aggravated disease. These findings correlated well with antigen-specific T cell proliferation and cytokine and antibody production. T cell proliferation was suppressed by the higher doses of CII, whereas the low dose enhanced T cell proliferation, indicating it primed the T cells. Suppression of T cell proliferation could be overcome by the addition of exogenous interleukin-2 (IL-2) to these cultures. Decreased T cell proliferation was associated with suppression of both Th1 (interferon-gamma [IFNgamma]) and Th2 (IL-4) cytokines and all the subclasses of anticollagen IgG in mice receiving 20, 80, or 320 microg of collagen. Overall, the highest dose of collagen (320 microg) was less effective at suppressing the immune response and disease than the 20-microg or 80-microg doses. There was an increased production of antibodies of all IgG isotypes, and of the Th1-associated cytokines IFNgamma and IL-2, in animals that had received the lowest dose of 5 microg collagen nasally. CONCLUSION Nasal administration of antigens is effective in inducing tolerance and reducing disease severity, but the effects are dose dependent. Low doses can prime the immune system and aggravate disease; high doses may not suppress disease. Suppression of the immune response, which correlates with suppression of disease, is not obviously associated with a type I to type II T cell switch, but rather with an overall suppression of both forms of T cell response, with a potential role for anergy of T cells in this process.
Collapse
Affiliation(s)
- C J Derry
- Division of Life Sciences, King's College London, UK
| | | | | | | | | |
Collapse
|