1
|
Zhong Y, Su C, Wu S, Miao C, Wang B. Nasal delivery of an immunotherapeutic vaccine in thermosensitive hydrogel against allergic asthma. Int Immunopharmacol 2023; 116:109718. [PMID: 36738673 DOI: 10.1016/j.intimp.2023.109718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/22/2022] [Accepted: 01/08/2023] [Indexed: 02/05/2023]
Abstract
Asthma poses a significant threat to public health, with an estimated burden of over 334 million people worldwide. Available treatments are often inadequate. We developed a thermo-sensitive hydrogel vaccine containing allergen and FK506 that induced immune tolerance via intranasal administration to treat experimental allergic asthma. The hydrogel delivery system was formulated based on Poloxamer 407 (P407), Carbopol 974P NF, and Polyoxyl 15 hydroxystearate (Kolliphor HS15, HS15). It flowed freely at room temperature and rapidly formed a hydrogel in the nasal cavity once the temperature rose over 33 °C. Ovalbumin and FK506 were slowly released from the hydrogel form and their mucosal residence time was significantly prolonged compared to the liquid formulation. In both an OVA-induced asthma model and an HDM-induced asthma model, the vaccines formulated in hydrogel gave lower levels of eosinophilic inflammation, and airway remodeling. The reduction of lung function was ameliorated, and Foxp3-expressing CD4 + Treg cells were significantly higher. The frequency of Foxp3 + Tregs in lung-draining lymph nodes (dLNs) was correlated with the amelioration. Depletion of Foxp3 + Treg cells abolished the beneficial effects of the allergen/FK506 hydrogel vaccinations. Thus, the allergen/FK506 hydrogel formulation has the potential to be a delivery system for therapeutic allergy vaccines to induce immune tolerance.
Collapse
Affiliation(s)
- Yiwei Zhong
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China; Fudan-Advaccine Join-Lab for Vaccine Research, Fudan University, Shanghai, China; Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Caixia Su
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China; Fudan-Advaccine Join-Lab for Vaccine Research, Fudan University, Shanghai, China; Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Shuting Wu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China; Fudan-Advaccine Join-Lab for Vaccine Research, Fudan University, Shanghai, China; Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Chunhui Miao
- Advaccine Biopharmaceutics (Suzhou) Co. Ltd, Suzhou, Jiangsu Province, China
| | - Bin Wang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China; Fudan-Advaccine Join-Lab for Vaccine Research, Fudan University, Shanghai, China; Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China; National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China; Children's Hospital of Fudan University, Shanghai, China.
| |
Collapse
|
2
|
Domínguez-Horta MDC, Serrano-Díaz A, Hernández-Cedeño M, Martínez-Donato G, Guillén-Nieto G. A peptide derived from HSP60 reduces proinflammatory cytokines and soluble mediators: a therapeutic approach to inflammation. Front Immunol 2023; 14:1162739. [PMID: 37187739 PMCID: PMC10179499 DOI: 10.3389/fimmu.2023.1162739] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 04/18/2023] [Indexed: 05/17/2023] Open
Abstract
Cytokines are secretion proteins that mediate and regulate immunity and inflammation. They are crucial in the progress of acute inflammatory diseases and autoimmunity. In fact, the inhibition of proinflammatory cytokines has been widely tested in the treatment of rheumatoid arthritis (RA). Some of these inhibitors have been used in the treatment of COVID-19 patients to improve survival rates. However, controlling the extent of inflammation with cytokine inhibitors is still a challenge because these molecules are redundant and pleiotropic. Here we review a novel therapeutic approach based on the use of the HSP60-derived Altered Peptide Ligand (APL) designed for RA and repositioned for the treatment of COVID-19 patients with hyperinflammation. HSP60 is a molecular chaperone found in all cells. It is involved in a wide diversity of cellular events including protein folding and trafficking. HSP60 concentration increases during cellular stress, for example inflammation. This protein has a dual role in immunity. Some HSP60-derived soluble epitopes induce inflammation, while others are immunoregulatory. Our HSP60-derived APL decreases the concentration of cytokines and induces the increase of FOXP3+ regulatory T cells (Treg) in various experimental systems. Furthermore, it decreases several cytokines and soluble mediators that are raised in RA, as well as decreases the excessive inflammatory response induced by SARS-CoV-2. This approach can be extended to other inflammatory diseases.
Collapse
Affiliation(s)
- Maria del Carmen Domínguez-Horta
- Autoimmunity Project, Pharmaceutical Division, Center for Genetic Engineering and Biotechnology, Havana, Cuba
- Physiology Department, Latin American School of Medicine, Havana, Cuba
- *Correspondence: Maria del Carmen Domínguez-Horta,
| | - Anabel Serrano-Díaz
- Autoimmunity Project, Pharmaceutical Division, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Mabel Hernández-Cedeño
- Autoimmunity Project, Pharmaceutical Division, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Gillian Martínez-Donato
- Biomedical Research Division, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| | - Gerardo Guillén-Nieto
- Physiology Department, Latin American School of Medicine, Havana, Cuba
- Biomedical Research Division, Center for Genetic Engineering and Biotechnology, Havana, Cuba
| |
Collapse
|
3
|
Peptide-Based Vaccination Therapy for Rheumatic Diseases. J Immunol Res 2020; 2020:8060375. [PMID: 32258176 PMCID: PMC7104265 DOI: 10.1155/2020/8060375] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 02/28/2020] [Indexed: 02/06/2023] Open
Abstract
Rheumatic diseases are extremely heterogeneous diseases with substantial risks of morbidity and mortality, and there is a pressing need in developing more safe and cost-effective treatment strategies. Peptide-based vaccination is a highly desirable strategy in treating noninfection diseases, such as cancer and autoimmune diseases, and has gained increasing attentions. This review is aimed at providing a brief overview of the recent advances in peptide-based vaccination therapy for rheumatic diseases. Tremendous efforts have been made to develop effective peptide-based vaccinations against rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE), while studies in other rheumatic diseases are still limited. Peptide-based active vaccination against pathogenic cytokines such as TNF-α and interferon-α (IFN-α) is shown to be promising in treating RA or SLE. Moreover, peptide-based tolerogenic vaccinations also have encouraging results in treating RA or SLE. However, most studies available now have been mainly based on animal models, while evidence from clinical studies is still lacking. The translation of these advances from experimental studies into clinical therapy remains impeded by some obstacles such as species difference in immunity, disease heterogeneity, and lack of safe delivery carriers or adjuvants. Nevertheless, advances in high-throughput technology, bioinformatics, and nanotechnology may help overcome these impediments and facilitate the successful development of peptide-based vaccination therapy for rheumatic diseases.
Collapse
|
4
|
Nasal route for vaccine and drug delivery: Features and current opportunities. Int J Pharm 2019; 572:118813. [PMID: 31678521 DOI: 10.1016/j.ijpharm.2019.118813] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 10/16/2019] [Accepted: 10/17/2019] [Indexed: 01/12/2023]
Abstract
Mucosal administration, and specifically nasal route, constitutes an alternative and promising strategy for drug and vaccine delivery. Mucosal routes have several advantages supporting their selective use for different pathologies. Currently, many efforts are being made to develop effective drug formulations and novel devices for nasal delivery. This review described the structure and main characteristics of the nasal cavity. The advantages, achievements and challenges of the nasal route use for medical purposes are discussed, with particular focus on vaccine delivery. Compelling evidences support the potentialities and safety of the nasal delivery of vaccines and drugs. This alternative route could become a solution for many unmet medical issues and also may facilitate and cheapen massive immunization campaigns or long-lasting chronic treatments. Nowadays, in spite of certain remaining skepticism, the field of nasal delivery of drugs and vaccines is growing fast, bolstered by current developments in nanotechnology, imaging and administration devices. A notable increase in the number of approved drugs for nasal administration is envisaged.
Collapse
|
5
|
Bernocchi B, Carpentier R, Betbeder D. Nasal nanovaccines. Int J Pharm 2017; 530:128-138. [PMID: 28698066 DOI: 10.1016/j.ijpharm.2017.07.012] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 07/03/2017] [Accepted: 07/04/2017] [Indexed: 01/08/2023]
Abstract
Nasal administration of vaccines is convenient for the potential stimulation of mucosal and systemic immune protection. Moreover the easy accessibility of the intranasal route renders it optimal for pandemic vaccination. Nanoparticles have been identified as ideal delivery systems and adjuvants for vaccine application. Heterogeneous protocols have been used for animal studies. This complicates the understanding of the formulation influence on the immune response and the comparison of the different nanoparticles approaches developed. Moreover anatomical and immunological differences between rodents and humans provide an additional hurdle in the rational development of nasal nanovaccines. This review will give a comprehensive expertise of the state of the art in nasal nanovaccines in animals and humans focusing on the nanomaterial used.
Collapse
Affiliation(s)
- B Bernocchi
- Inserm, LIRIC-UMR 995, F-59000 Lille, France; Université de Lille, LIRIC-UMR 995, F-59000 Lille, France; CHRU de Lille, LIRIC-UMR 995, F-59000 Lille, France
| | - R Carpentier
- Inserm, LIRIC-UMR 995, F-59000 Lille, France; Université de Lille, LIRIC-UMR 995, F-59000 Lille, France; CHRU de Lille, LIRIC-UMR 995, F-59000 Lille, France.
| | - D Betbeder
- Inserm, LIRIC-UMR 995, F-59000 Lille, France; Université de Lille, LIRIC-UMR 995, F-59000 Lille, France; CHRU de Lille, LIRIC-UMR 995, F-59000 Lille, France; University of Artois, 62000 Arras, France
| |
Collapse
|
6
|
Spierings J, van Eden W. Heat shock proteins and their immunomodulatory role in inflammatory arthritis. Rheumatology (Oxford) 2016; 56:198-208. [PMID: 27411479 DOI: 10.1093/rheumatology/kew266] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Revised: 05/24/2016] [Indexed: 11/14/2022] Open
Abstract
Autoimmune diseases, including inflammatory arthritis, are characterized by a loss of self-tolerance, leading to an excessive immune responses and subsequent ongoing inflammation. Current therapies are focused on dampening this inflammation, but a permanent state of tolerance is seldom achieved. Therefore, novel therapies that restore and maintain tolerance are needed. Tregs could be a potential target to achieve permanent immunotolerance. Activation of Tregs can be accomplished when they recognize and bind their specific antigens. HSPs are proteins present in all cells and are upregulated during inflammation. These proteins are immunogenic and can be recognized by Tregs. Several studies in animal models and in human clinical trials have shown the immunoregulatory effects of HSPs and their protective effects in inflammatory arthritis. In this review, an overview is presented of the immunomodulatory effects of several members of the HSP family in general and in inflammatory arthritis. These effects can be attributed to the activation of Tregs through cellular interactions within the immune system. The effect of HSP-specific therapies in patients with inflammatory arthritis should be explored further, especially with regard to long-term efficacy and safety and their use in combination with current therapeutic approaches.
Collapse
Affiliation(s)
- Julia Spierings
- Department of Rheumatology, Maastricht University Medical Center, Maastricht
| | - Willem van Eden
- Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
7
|
Affiliation(s)
- Bernardo Rodriguez-Iturbe
- From the Department of Nephrology, Hospital Universitario and Instituto Venezolano de Investigaciones Científicas (IVIC)-Zulia, Maracaibo, Venezuela.
| |
Collapse
|
8
|
Shi XL, Wang LP, Feng X, Fan DD, Zang WJ, Wang B, Zhou J. Inhibition of adjuvant-induced arthritis by nasal administration of novel synthetic peptides from heat shock protein 65. BMC Musculoskelet Disord 2014; 15:253. [PMID: 25059987 PMCID: PMC4115216 DOI: 10.1186/1471-2474-15-253] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Accepted: 07/18/2014] [Indexed: 12/17/2022] Open
Abstract
Background Rheumatoid arthritis (RA) is a chronic systemic inflammatory disease mediated by T cells. The aim of the present study was to investigate the therapeutic efficacy of synthetic peptides (HP-R1, HP-R2 and HP-R3), derived from the sequence of 65-kD mycobacterial heat shock protein (HSP), in the treatment of RA using adjuvant-induced arthritis (AA) animal model. Methods AA was induced by a single intradermal injection Freund’s complete adjuvant in male Lewis rats. At the first clinical sign of disease, rats were administered nasally by micropipette of peptides or phosphate buffer saline (PBS). Disease progression was monitored by measurement of body weight, arthritis score and paw swelling. The changes of histopathology were assessed by hematoxylin eosin staining. The serum levels of tumor necrosis factor (TNF) - alpha and interleukin (IL)-4 were measured by enzyme-linked immunosorbent assay (ELISA). Results The peptides efficiently inhibited the footpad swelling and arthritic symptoms in AA rats. The synthetic peptides displayed significantly less inflammatory cellular infiltration and synovium hyperplasia than model controls. This effect was associated with a suppression of pro-inflammatory cytokine TNF-alpha production and an increase of anti-inflammatory cytokine IL-4 production after peptides treatment. Conclusions These results suggest that the synthetic peptides derived from HSP65 induce highly effective protection against AA, which is mediated in part by down-regulation of inflammatory cytokines, and support the view that the synthetic peptides is a potential therapy for RA that may help to diminish both joint inflammation and destruction.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jun Zhou
- Department of Pharmacology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, P,R, China.
| |
Collapse
|
9
|
Abstract
The T-cell component of the antigen-specific immune response is the target of various novel interventions to modify chronic immunologic disorders, such as allergic diseases. Recent clinical trials have evaluated the safety and efficacy of therapeutic vaccines consisting of short, synthetic, allergen-derived peptides, corresponding to T-cell epitopes from the eliciting antigen. The main advantage of such an approach is the reduction in systemic, immunoglobulin E-mediated adverse events compared with existing whole allergen immunotherapy, often referred to as 'allergy shots'. T-cell peptide epitopes, although capable of inducing immunologic tolerance, are short linear structures that have reduced ability to cross-link mast cell- and basophil-bound immunoglobulin E. The precise mechanism of tolerance induction remains incompletely defined. However, recent data indicate that peptide therapy induces/expands a population of antigen-specific regulatory T-cells. A novel form of treatment combining efficacy with a substantially decreased occurrence of adverse events is likely to have a major impact on the management and prevalence of allergic diseases. Furthermore, the principles of epitope-specific therapy hold promise for the development of therapeutic vaccines for the treatment of autoimmune diseases.
Collapse
Affiliation(s)
- F Runa Ali
- Faculty of Medicine, Imperial College, Department of Allergy and Clinical Immunology, Respiratory Immunology Group,Rm 360, Sir Alexander Fleming Building,Faculty of Medicine, Imperial College, London SW7 2AZ, UK
| | | |
Collapse
|
10
|
Cappello F, Marino Gammazza A, Palumbo Piccionello A, Campanella C, Pace A, Conway de Macario E, Macario AJL. Hsp60 chaperonopathies and chaperonotherapy: targets and agents. Expert Opin Ther Targets 2013; 18:185-208. [PMID: 24286280 DOI: 10.1517/14728222.2014.856417] [Citation(s) in RCA: 103] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Hsp60 (Cpn60) assembles into a tetradecamer that interacts with the co-chaperonin Hsp10 (Cpn10) to assist client polypeptides to fold, but it also has other roles, including participation in pathogenic mechanisms. AREA COVERED Hsp60 chaperonopathies are pathological conditions, inherited or acquired, in which the chaperone plays a determinant etiologic-pathogenic role. These diseases justify selection of Hsp60 as a target for developing agents that interfere with its pathogenic effects. We provide information on how to proceed. EXPERT OPINION The information available encourages the development of ways to improve Hsp60 activity (positive chaperonotherapy) when deficient or to block it (negative chaperonotherapy) when pathogenic. Many questions are still unanswered and obstacles are obvious. More information is needed to establish when and why autologous Hsp60 becomes a pathogenic autoantigen, or induces cytokine formation and inflammation, or favors carcinogenesis. Clarification of these points will take considerable time. However, analysis of the Hsp60 molecule and a search for active compounds aimed at structural sites that will affect its functioning should continue without interruption. No doubt that some of these compounds will offer therapeutic hopes and will also be instrumental for dissecting structure-function relationships at the biochemical and biological (using animal models and cultured cells) levels.
Collapse
Affiliation(s)
- Francesco Cappello
- Euro-Mediterranean Institute of Science and Technology (IEMEST) , Palermo , Italy
| | | | | | | | | | | | | |
Collapse
|
11
|
Mackenzie KJ, Fitch PM, Leech MD, Ilchmann A, Wilson C, McFarlane AJ, Howie SEM, Anderton SM, Schwarze J. Combination peptide immunotherapy based on T-cell epitope mapping reduces allergen-specific IgE and eosinophilia in allergic airway inflammation. Immunology 2013; 138:258-68. [PMID: 23113712 DOI: 10.1111/imm.12032] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2012] [Revised: 10/16/2012] [Accepted: 10/22/2012] [Indexed: 01/04/2023] Open
Abstract
Peptide immunotherapy using soluble peptides containing allergen-derived immunodominant T-cell epitopes holds therapeutic promise for allergic asthma. Previous studies in BALB/c mice using the immunodominant peptide epitope of chicken ovalbumin (p323-339) have been unable to demonstrate therapeutic effects in ovalbumin-induced allergic airway inflammation. We have previously shown that intravenous application of p323-339 can effectively tolerise p323-339-reactive T cells in a non-allergic model in C57BL/6 mice. This study aimed to assess the effects of using p323-339 immunotherapy in a C57BL/6 model of ovalbumin-induced allergic airway inflammation, identify any additional epitopes recognized by the ovalbumin-responsive T-cell repertoire in C57BL/6 mice and assess the effects of combination peptide immunotherapy in this model. Ovalbumin-reactive T-cell lines were generated from ovalbumin-immunized C57BL/6 mice and proliferative responses to a panel of overlapping peptides covering the ovalbumin sequence were assessed. Soluble peptides (singly or combined) were administered intravenously to C57BL/6 mice before the induction of ovalbumin-induced allergic airway inflammation. Peptide immunotherapy using the 323-339 peptide alone did not reduce the severity of allergic airway inflammation. An additional immunodominant T-cell epitope in ovalbumin was identified within the 263-278 sequence. Combination peptide immunotherapy, using the 323-339 and 263-278 peptides together, reduced eosinophilia in the bronchoalveolar lavage and ovalbumin-specific IgE, with apparent reductions in interleukin-5 and interleukin-13. Characterization of the T-cell response to a model allergen has allowed the development of combination peptide immunotherapy with improved efficacy in allergic airway inflammation. This model holds important potential for future mechanistic studies using peptide immunotherapy in allergy.
Collapse
Affiliation(s)
- Karen J Mackenzie
- MRC Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Zonneveld-Huijssoon E, van Wijk F, Roord S, Delemarre E, Meerding J, de Jager W, Klein M, Raz E, Albani S, Kuis W, Boes M, Prakken BJ. TLR9 agonist CpG enhances protective nasal HSP60 peptide vaccine efficacy in experimental autoimmune arthritis. Ann Rheum Dis 2012; 71:1706-15. [PMID: 22562976 DOI: 10.1136/annrheumdis-2011-201131] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
OBJECTIVES Peptide-based immune tolerance induction is considered an attractive treatment option for autoimmune diseases. The authors have developed a novel method that can enhance the induction of protective peptide-specific T-cell responses, using a rat arthritis model. The authors focused on the Toll-like receptor 9 ligand CpG, which was shown to stimulate regulatory T-cell proliferation when added to plasmacytoid dendritic cells (pDC) using in-vitro cultures. METHODS The peptide used is a heat shock protein 60 epitope (p1) that elicits tolerogenic peptide-specific immune responses in human arthritis patients and was recently shown to have protective capacity as a bystander antigen in the rat adjuvant arthritis model. Rats were treated with three nasal doses of p1, CpG or a combination of p1 and CpG. Antigen-presenting cells were studied in nose-draining lymph nodes (mandibular lymph nodes; MLN) after nasal treatment, and T-cell responses were analysed in joint-draining lymph nodes after arthritis induction. RESULTS Nasal co-administration of p1/CpG significantly augmented the arthritis-protective effect of p1, while CpG treatment alone did not. Co-treatment of p1/CpG increased both the number and activation status of pDC in draining MLN, which was accompanied by amplified p1-specific T-cell proliferation and interleukin (IL)-10 production. During early arthritis, p1-specific IL-10 production was identified at the site of inflammation. P1 and p1/CpG-treated rats showed a greater amount of CD4+FoxP3+ regulatory T cells in the joint-draining lymph nodes, which correlated with lower arthritis scores. CONCLUSIONS These clinical and immunological data suggest the use of CpG as a potent adjuvant for mucosal peptide-specific immune therapy in arthritis.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Administration, Intranasal
- Animals
- Arthritis, Experimental/immunology
- Arthritis, Rheumatoid/immunology
- Chaperonin 60/administration & dosage
- Chaperonin 60/immunology
- Dendritic Cells/immunology
- Disease Models, Animal
- Epitopes, T-Lymphocyte/immunology
- Lymphocyte Activation/immunology
- Male
- Oligodeoxyribonucleotides/administration & dosage
- Oligodeoxyribonucleotides/immunology
- Rats
- Rats, Inbred Lew
- T-Lymphocytes, Regulatory/immunology
- Toll-Like Receptor 9/agonists
- Vaccines, Subunit/administration & dosage
- Vaccines, Subunit/immunology
Collapse
Affiliation(s)
- Evelien Zonneveld-Huijssoon
- Department of Pediatric Immunology, University Medical Centre Utrecht, Centre for Molecular and Cellular Intervention, Lundlaan 6, 3584 EA Utrecht, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Keijzer C, Wieten L, van Herwijnen M, van der Zee R, Van Eden W, Broere F. Heat shock proteins are therapeutic targets in autoimmune diseases and other chronic inflammatory conditions. Expert Opin Ther Targets 2012; 16:849-57. [PMID: 22793002 DOI: 10.1517/14728222.2012.706605] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Exploitation of antigen-specific regulatory T cells (Tregs) as critical regulators in the control of chronic inflammatory diseases is hampered by the obscure nature of most disease-relevant autoantigens. Heat shock proteins (Hsp) are possible targets for Tregs due to their enhanced expression in inflamed (stressed) tissues and there is evidence that Hsp can induce anti-inflammatory immunoregulatory T-cell responses. AREAS COVERED Recent publications showing that exogenous administration of stress proteins has induced immunoregulation in various models of inflammatory disease have also been shown to be effective in first clinical trials in humans. Now, in the light of a growing interest in T-cell regulation, it is of interest to further explore the mechanisms through which Hsp can be utilized to trigger immunoregulatory pathways, capable of suppressing such a wide and diversified spectrum of inflammatory diseases. EXPERT OPINION Therapeutic approaches via exploitation of antigen-specific Tregs will benefit from tailor-made combination therapies. Combining current therapeutic approaches with Hsp-specific therapies thereby enhancing natural immune regulation might expedite the entry of antigen-specific regulatory T cells into the therapeutic arsenal of the anti-inflammatory therapeutics.
Collapse
Affiliation(s)
- Chantal Keijzer
- University Utrecht, Faculty Veterinary Medicine, Department Infectious Diseases and Immunology, Yalelaan, Netherlands
| | | | | | | | | | | |
Collapse
|
14
|
Coelho V, Faria AMC. HSP60: issues and insights on its therapeutic use as an immunoregulatory agent. Front Immunol 2012; 2:97. [PMID: 22566886 PMCID: PMC3342027 DOI: 10.3389/fimmu.2011.00097] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2011] [Accepted: 12/28/2011] [Indexed: 01/05/2023] Open
Abstract
Heat shock proteins 60 (HSP60) is one of the most well studied member of the HSP family. Although found to be a target self antigen in pathological autoimmunity and HSP60-reactive T and B cells are part of immune responses in several infectious diseases, there is consistent experimental evidence that HSP60 displays dominant immunoregulatory properties. There are a series of reports on animal models showing that the administration of HSP60 can modulate inflammatory diseases. However, HSP60 has both immune-regulatory and inflammatory properties placing it as an essentially homeostatic antigen, but with potentially harmful effects as well. There have been a series of reports on the successful use of HSP60 and its peptides as immune-modulatory agent for several models of autoimmune diseases and in some clinical trials as well. We believe that the potential risks of HSP60 as a therapeutic agent can be controlled by addressing important factors determining its effects. These factors would be route of administration, appropriate peptides, time point of administration in the course of the disease, and possible association with other modulatory agents.
Collapse
Affiliation(s)
- Verônica Coelho
- Laboratório de Imunologia, Instituto do Coração, Universidade de São Paulo São Paulo, Brazil
| | | |
Collapse
|
15
|
Zonneveld-Huijssoon E, Roord STA, de Jager W, Klein M, Albani S, Anderton SM, Kuis W, van Wijk F, Prakken BJ. Bystander suppression of experimental arthritis by nasal administration of a heat shock protein peptide. Ann Rheum Dis 2011; 70:2199-206. [PMID: 21914624 DOI: 10.1136/ard.2010.136994] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
OBJECTIVES Mucosal immune therapy with disease-inducing antigens is an effective way to prevent experimental arthritis, but in humans these antigens are unknown. In juvenile idiopathic arthritis, however, T cell recognition of a so-called bystander antigen, heat shock protein 60 (HSP60), is associated with a good prognosis. Recently epitopes derived from HSP60, a microbial peptide (p1) and its self-homologue (p2) were reported to induce tolerogenic T cell responses in vitro in patients with arthritis. A study was undertaken to determine whether mucosal administration of these bystander epitopes can be similarly effective in suppressing arthritis. METHODS Rats were treated nasally with p1, p2 or phosphate-buffered saline before arthritis induction. Arthritis scores were assessed and peptide-specific proliferative responses, phenotypic analysis, cytokine production and in vitro suppressive capacity of cells were measured in lymph nodes and spleens. CD4 spleen T cells from p1- or p2-treated rats were adoptively transferred into naïve rats that were subsequently injected with complete Freund's adjuvant for arthritis induction. RESULTS Nasal administration of p1 prevented experimental arthritis whereas treatment with the self-homologue p2 did not. Adoptive transfer of CD4 T cells protected against experimental arthritis. Treatment with p1 increased peptide-specific and self-crossreactive interferon γ (IFNγ) production. Tumour necrosis factor α (TNFα) levels were reduced at the site of inflammation. Forkhead box P3 (FoxP3) expression remained stable but the suppressive capacity of T regulatory cells in p1-treated rats was enhanced. CONCLUSION p1 immune therapy induces a population of CD4 T cells with reduced TNFα and increased peptide-specific IFNγ production at the site of inflammation. This population expresses FoxP3 and has potent suppressive capacity which, upon transfer, protects against arthritis. The bystander epitope p1 may therefore be a suitable candidate for antigen-specific immunotherapy in arthritis.
Collapse
MESH Headings
- Administration, Intranasal
- Animals
- Arthritis, Experimental/immunology
- Arthritis, Experimental/prevention & control
- Bystander Effect/immunology
- CD4-Positive T-Lymphocytes/transplantation
- Chaperonin 60/administration & dosage
- Chaperonin 60/immunology
- Chaperonin 60/therapeutic use
- Cytokines/biosynthesis
- Epitopes, T-Lymphocyte/administration & dosage
- Epitopes, T-Lymphocyte/immunology
- Epitopes, T-Lymphocyte/therapeutic use
- Freund's Adjuvant
- Immunity, Mucosal
- Immunotherapy, Adoptive/methods
- Inflammation Mediators/metabolism
- Lymphocyte Activation/immunology
- Male
- Peptide Fragments/administration & dosage
- Peptide Fragments/immunology
- Peptide Fragments/therapeutic use
- Rats
- Rats, Inbred Lew
- Spleen/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes, Regulatory/immunology
Collapse
Affiliation(s)
- Evelien Zonneveld-Huijssoon
- Department of Pediatric Immunology, Centre for Molecular and Cellular Intervention, University Medical Centre Utrecht, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Yu H, Lu C, Tan MT, Moudgil KD. The gene expression profile of preclinical autoimmune arthritis and its modulation by a tolerogenic disease-protective antigenic challenge. Arthritis Res Ther 2011; 13:R143. [PMID: 21914168 PMCID: PMC3308071 DOI: 10.1186/ar3457] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2011] [Revised: 06/29/2011] [Accepted: 09/13/2011] [Indexed: 02/06/2023] Open
Abstract
Introduction Autoimmune inflammation is a characteristic feature of rheumatoid arthritis (RA) and other autoimmune diseases. In the natural course of human autoimmune diseases, it is rather difficult to pinpoint the precise timing of the initial event that triggers the cascade of pathogenic events that later culminate into clinically overt disease. Therefore, it is a challenge to examine the early preclinical events in these disorders. Animal models are an invaluable resource in this regard. Furthermore, considering the complex nature of the pathogenic immune events in arthritis, microarray analysis offers a versatile tool to define the dynamic patterns of gene expression during the disease course. Methods In this study, we defined the profiles of gene expression at different phases of adjuvant arthritis (AA) in Lewis rats and compared them with those of antigen mycobacterial heat shock protein 65 (Bhsp65)-tolerized syngeneic rats. Purified total RNA (100 ng) extracted from the draining lymph node cells was used to generate biotin-labeled fragment cRNA, which was then hybridized with an oligonucleotide-based DNA microarray chip. Significance analysis of microarrays was used to compare gene expression levels between the two different groups by limiting the false discovery rate to < 5%. Some of the data were further analyzed using a fold change ≥2.0 as the cutoff. The gene expression of select genes was validated by quantitative real-time PCR. Results Intriguingly, the most dramatic changes in gene expression in the draining lymphoid tissue ex vivo were observed at the preclinical (incubation) phase of the disease. The affected genes represented many of the known proteins that participate in the cellular immune response. Interestingly, the preclinical gene expression profile was significantly altered by a disease-modulating, antigen-based tolerogenic regimen. The changes mostly included upregulation of several genes, suggesting that immune tolerance suppressed disease by activating disease-regulating pathways. We identified a molecular signature comprising at least 12 arthritis-related genes altered by Bhsp65-induced tolerance. Conclusions This is the first report of microarray analysis in the rat AA model. The results of this study not only advance our understanding of the early phase events in autoimmune arthritis but also help in identifying potential targets for the immunomodulation of RA.
Collapse
Affiliation(s)
- Hua Yu
- Department of Microbiology and Immunology, University of Maryland School of Medicine, 685 West Baltimore Street, HSF-1, Suite 380, Baltimore, MD 21201, USA
| | | | | | | |
Collapse
|
17
|
Abstract
Specific allergen immunotherapy is clinically effective and disease modifying. It has a duration of effect that exceeds the treatment period and prevents both the progression of allergic rhinitis to asthma and the acquisition of new allergic sensitizations. However, immunotherapy is associated with a high frequency of adverse events related to the allergenicity of vaccines. Allergenicity is conferred by the presence of intact B-cell epitopes that crosslink allergen-specific IgE on effector cells. The use of linear peptide sequences representing fragments of the native allergen is one approach to reduce allergenicity. Preclinical models of peptide immunotherapy have demonstrated efficacy in both autoimmunity and allergy. Translation of this technology into the clinic has gained momentum in recent years based on encouraging results from early clinical trials. To date, efforts have focused on two major allergens, but vaccines to a broader range of molecules are currently in clinical development. Mechanistically, peptide immunotherapy appears to work through the induction of adaptive, allergen-specific regulatory T cells that secrete the immunoregulatory cytokine IL-10. There is also evidence that peptide immunotherapy targeting allergen-specific T cells can indirectly modulate allergen-specific B-cell responses. Peptide immunotherapy may provide a safe and efficacious alternative to conventional subcutaneous and/or sublingual approaches using native allergen preparations.
Collapse
Affiliation(s)
- D Moldaver
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, Hamilton, ON, Canada
| | | |
Collapse
|
18
|
Abstract
Specific immunotherapy (SIT) with extracts containing intact allergen molecules is clinically efficacious, but associated with frequent adverse events related to the allergic sensitization of the patient. As a result, treatment is initiated in an incremental dose fashion which ultimately achieves a plateau (maintenance dose) that may be continued for several years. Reduction of allergic adverse events may allow safer and more rapid treatment Thus, many groups have developed and evaluated strategies to reduce allergenicity whilst maintaining immunogenicity, the latter being required to achieve specific modulation of the immune response. Peptide immunotherapy can be used to target T and/or B cells in an antigen-specific manner. To date, only approaches that target T cells have been clinically evaluated. Short, synthetic peptides representing immunodominant T cell epitopes of major allergens are able to modulate allergen-specific T cell responses in the absence of IgE cross linking and activation of effector cells. Here we review clinical and mechanistic studies associated with peptide immunotherapy targeting allergy to cats or to bee venom.
Collapse
Affiliation(s)
- Mark Larché
- Department of Medicine, Firestone Institute for Respiratory Health, McMaster University, HSC 4H20, Hamilton, ON L8N 3Z5, Canada.
| |
Collapse
|
19
|
Gwinn WM, Kirwan SM, Wang SH, Ashcraft KA, Sparks NL, Doil CR, Tlusty TG, Casey LS, Hollingshead SK, Briles DE, Dondero RS, Hickey AJ, Foster WM, Staats HF. Effective induction of protective systemic immunity with nasally administered vaccines adjuvanted with IL-1. Vaccine 2010; 28:6901-14. [PMID: 20723629 DOI: 10.1016/j.vaccine.2010.08.006] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2010] [Revised: 07/23/2010] [Accepted: 08/02/2010] [Indexed: 11/26/2022]
Abstract
IL-1α and IL-1β were evaluated for their ability to provide adjuvant activity for the induction of serum antibody responses when nasally administered with protein antigens in mice and rabbits. In mice, intranasal (i.n.) immunization with pneumococcal surface protein A (PspA) or tetanus toxoid (TT) combined with IL-1β induced protective immunity that was equivalent to that induced by parenteral immunization. Nasal immunization of awake (i.e., not anesthetized) rabbits with IL-1-adjuvanted vaccines induced highly variable serum antibody responses and was not as effective as parenteral immunization for the induction of antigen-specific serum IgG. However, i.n. immunization of deeply anesthetized rabbits with rPA+IL-1α consistently induced rPA-specific serum IgG ELISA titers that were not significantly different than those induced by intramuscular (IM) immunization with rPA+alum although lethal toxin-neutralizing titers induced by nasal immunization were lower than those induced by IM immunization. Gamma scintigraphy demonstrated that the enhanced immunogenicity of nasal immunization in anesthetized rabbits correlated with an increased nasal retention of i.n. delivered non-permeable radio-labeled colloidal particles. Our results demonstrate that, in mice, IL-1 is an effective adjuvant for nasally administered vaccines for the induction of protective systemic immunity and that in non-rodent species, effective induction of systemic immunity with nasally administered vaccines may require formulations that ensure adequate retention of the vaccine within the nasal cavity.
Collapse
Affiliation(s)
- William M Gwinn
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
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: 0.7] [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
|
21
|
The involvement of heat-shock proteins in the pathogenesis of autoimmune arthritis: a critical appraisal. Semin Arthritis Rheum 2009; 40:164-75. [PMID: 19969325 DOI: 10.1016/j.semarthrit.2009.10.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2009] [Revised: 09/23/2009] [Accepted: 10/04/2009] [Indexed: 01/31/2023]
Abstract
OBJECTIVES To review the literature on the role of heat-shock proteins (HSPs) in the pathogenesis of autoimmune arthritis in animal models and patients with rheumatoid arthritis (RA). METHODS The published literature in Medline (PubMed), including our published work on the cell-mediated as well as humoral immune response to various HSPs, was reviewed. Studies in the preclinical animal models of arthritis as well as RA were examined critically and the data are presented. RESULTS In experimental arthritis, disease induction by different arthritogenic stimuli, including an adjuvant, led to immune response to mycobacterial HSP65 (BHSP65). However, attempts to induce arthritis by a purified HSP have not met with success. There are several reports of a significant immune response to HSP65 in RA patients. However, the issue of cause and effect is difficult to address. Nevertheless, several studies in animal models and a couple of clinical trials in RA patients have shown the beneficial effect of HSPs against autoimmune arthritis. CONCLUSIONS There is a clear association between immune response to HSPs, particularly HSP65, and the initiation and propagation of autoimmune arthritis in experimental models. The correlation is relatively less convincing in RA patients. In both cases, the ability of HSPs to modulate arthritis offers support, albeit an indirect one, for the involvement of these antigens in the disease process.
Collapse
|
22
|
Durai M, Huang MN, Moudgil KD. Self heat-shock protein 65-mediated regulation of autoimmune arthritis. J Autoimmun 2009; 33:208-13. [PMID: 19800761 DOI: 10.1016/j.jaut.2009.09.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Heat-shock proteins (Hsps) have been invoked in the pathogenesis of a variety of autoimmune diseases. The mycobacterial heat-shock protein 65 (Bhsp65) has been studied extensively as one of the antigenic triggers of autoimmunity in experimental models of, as well as patients with, rheumatoid arthritis. As Hsps are highly conserved and immunogenic, it is generally anticipated that self Hsps might serve as the endogenous targets of the immune response initiated by the homologous foreign Hsps. Contrary to this expectation, studies in the rat adjuvant arthritis (AA) model have revealed that priming of the self (rat) hsp65 (Rhsp65)-directed T cells in the Lewis rat leads to protection against AA instead of disease induction or aggravation. The arthritis-protective attribute of the self hsp65 is also evident following spontaneous priming of the anti-Rhsp65 T cells during the natural course of AA. Furthermore, immunization of rats with human hsp60, or with Bhsp65 peptides that are crossreactive with the corresponding self hsp65 peptides, leads to protection against AA. Importantly, high levels of T cell reactivity against self hsp60 in patients with juvenile idiopathic arthritis positively correlate with a favorable outcome of the disease. Thus, immune response against self hsp65 in autoimmune arthritis is protective rather than being pathogenic.
Collapse
Affiliation(s)
- Malarvizhi Durai
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Howard Hall Room 323 C, 660 West Redwood St., Baltimore, MD 21201, USA
| | | | | |
Collapse
|
23
|
Moudgil KD, Durai M. Regulation of autoimmune arthritis by self-heat-shock proteins. Trends Immunol 2009; 29:412-8. [PMID: 18675587 DOI: 10.1016/j.it.2008.06.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2008] [Revised: 06/20/2008] [Accepted: 06/26/2008] [Indexed: 12/17/2022]
Abstract
Heat-shock proteins (hsps) are highly conserved and immunogenic, and they are generally perceived to be attractive initiators or targets of a pathogenic immune response, and as such, have been implicated in the pathogenesis of autoimmune arthritis. However, studies in animal models and arthritis patients have unraveled the disease-regulating attributes of self-hsp65. We propose that the self-hsp65 induces a protective and beneficial immune response because of its ubiquitous distribution, stress inducibility and participation in tolerogenic processes. By contrast, the foreign hsp65 that does not influence the above processes and that resides admixed with microbial ligands for innate receptors generates an inflammatory pathogenic response. The regulatory properties of self-hsps need be fully explored and might be used for therapeutic purposes.
Collapse
Affiliation(s)
- Kamal D Moudgil
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | | |
Collapse
|
24
|
Root-Bernstein R. A measles-derived peptide treats and vaccinates against adjuvant arthritis. Autoimmun Rev 2009; 8:405-9. [PMID: 19124087 DOI: 10.1016/j.autrev.2006.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2006] [Accepted: 03/12/2006] [Indexed: 11/16/2022]
Abstract
Measles vaccine and porcine myelin basic protein were both found to ameliorate or abolish the symptoms of adjuvant arthritis (AA) in Lewis rats whether inoculated at the time of adjuvant administration or after the onset of arthritis. These results are consistent with clinical observations that measles infection can sometimes cause remission of juvenile rheumatoid arthritis. The fact that measles virus proteins and myelin basic protein have significant regions of homology allowed peptides based on these regions to be synthesized. A twenty-amino acid sequence exhibits significant anti-arthritic activity when inoculated into rats with pre-existing AA and it also prevented onset of AA when a single dose was preinoculated three weeks prior to AA induction. These data suggest the possibility of developing novel therapeutic vaccines against some forms of arthritis.
Collapse
|
25
|
Satpute SR, Durai M, Moudgil KD. Antigen-specific tolerogenic and immunomodulatory strategies for the treatment of autoimmune arthritis. Semin Arthritis Rheum 2008; 38:195-207. [PMID: 18177689 PMCID: PMC2723747 DOI: 10.1016/j.semarthrit.2007.10.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2007] [Revised: 10/18/2007] [Accepted: 10/21/2007] [Indexed: 12/15/2022]
Abstract
OBJECTIVES To review various antigen-specific tolerogenic and immunomodulatory approaches for arthritis in animal models and patients in regard to their efficacy, mechanisms of action, and limitations. METHODS We reviewed the published literature in Medline (PubMed) on the induction of antigen-specific tolerance and its effect on autoimmune arthritis, as well as the recent work on B-cell-mediated tolerance from our laboratory. The prominent key words used in different combinations included arthritis, autoimmunity, immunotherapy, innate immunity, tolerance, treatment, and rheumatoid arthritis (RA). Although this search spanned the years 1975 to 2007, the majority of the short-listed articles belonged to the period 1990 to 2007. The relevant primary as well as cross-referenced articles were then collected from links within PubMed and reviewed. RESULTS Antigen-specific tolerance has been successful in the prevention and/or treatment of arthritis in animal models. The administration of soluble native antigen or an altered peptide ligand intravenously, orally, or nasally, and the delivery of the DNA encoding a particular antigen by gene therapy have been the mainstay of immunomodulation. Recently, the methods for in vitro expansion of CD4+CD25+ regulatory T-cells have been optimized. Furthermore, interleukin-17 has emerged as a promising new therapeutic target in arthritis. However, in RA patients, non-antigen-specific therapeutic approaches have been much more successful than antigen-specific tolerogenic regimens. CONCLUSION An antigen-specific treatment against autoimmune arthritis is still elusive. However, insights into newly emerging mechanisms of disease pathogenesis provide hope for the development of effective and safe immunotherapeutic strategies in the near future.
Collapse
Affiliation(s)
- Shailesh R. Satpute
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Malarvizhi Durai
- Johns Hopkins Medical Institutions, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Kamal D. Moudgil
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Division of Rheumatology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
26
|
Wick MC, Mayerl C, Backovic A, van der Zee R, Jaschke W, Dietrich H, Wick G. In vivo imaging of the effect of LPS on arterial endothelial cells: molecular imaging of heat shock protein 60 expression. Cell Stress Chaperones 2008; 13:275-85. [PMID: 18465205 PMCID: PMC2673942 DOI: 10.1007/s12192-008-0044-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2008] [Revised: 03/26/2008] [Accepted: 03/31/2008] [Indexed: 12/01/2022] Open
Abstract
Bacterial endotoxins are known as stress factors for endothelial cells. In 20 normocholesterolemic New Zealand White (NZW) rabbits, endothelial stress was induced by intravenous (i.v.) injection of lipopolysaccharide (LPS), while eight NZW rabbits were sham-treated or served as untreated controls. In vivo molecular imaging was performed using co-registered computer tomography and positron emission tomography 24 h after i.v. injection of (124)I-labeled monoclonal anti-HSP60 or (124)I-radiolabelled isotype control antibodies. Compared to control animals, in vivo images of rabbit aortae revealed significantly increased endothelial binding of (124)I-labeled anti-HSP60 antibodies upon LPS, especially at sites of aortal branching. This was confirmed by immunohistochemistry and autoradiography data. Our results showed, as proof-of-principle, that HSP60-expression in normocholesterolemic rabbits is significantly increased after induction of endothelial stress and that non-invasive in vivo molecular imaging of early aortal HSP60-expression using (124)I-labeled anti-HSP60 monoclonal antibodies is possible.
Collapse
Affiliation(s)
- Marius C Wick
- Department of Radiology, Innsbruck Medical University, Anichstrasse 35, Innsbruck, Austria.
| | | | | | | | | | | | | |
Collapse
|
27
|
Exogenous tumour necrosis factor alpha induces suppression of autoimmune arthritis. Arthritis Res Ther 2008; 10:R38. [PMID: 18380898 PMCID: PMC3386491 DOI: 10.1186/ar2393] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2008] [Revised: 03/12/2008] [Accepted: 04/01/2008] [Indexed: 11/10/2022] Open
Abstract
Introduction Our previous studies showed that arthritic Lewis (LEW) rats produced the highest levels of tumour necrosis factor (TNF)α in the recovery phase of adjuvant arthritis (AA), suggesting a correlation between high TNFα levels and reduced severity of arthritis. To further explore this correlation, we compared the TNFα secretion profile of the AA-resistant Wistar Kyoto (WKY) rats with that of LEW rats, determined the effect of exogenous TNFα on the course of AA in LEW rats, and examined various mechanisms involved in TNFα-induced disease modulation. Methods A cohort each of LEW and WKY rats was immunised subcutaneously with heat-killed Mycobacterium tuberculosis H37Ra (Mtb). At different time points thereafter, subgroups of rats were killed and their draining lymph node cells were tested for cytokine production. Another group of LEW rats was injected with TNFα intraperitoneally daily for a total of 10 injections, 3 before and 6 after Mtb challenge, and then observed for signs of AA. In parallel, TNFα-treated rats were examined for changes in other cytokines, in CD4+CD25+ T cell frequency, and in indoleamine 2,3-dioxygenase (IDO) mRNA expression levels. Results LEW rats displayed a TNFα secretion profile that was opposite to that of the WKY rats. Furthermore, TNFα treatment significantly downmodulated the severity of AA in LEW rats, and decreased the interferon (IFN)-γ secretion in response to the pathogenic determinant of the disease-related antigen. No significant alterations were observed in other parameters tested. Conclusion The role of endogenous TNFα in the induction and propagation of arthritis is well established. However, exogenous TNFα can downmodulate the course of AA, displaying an immunoregulatory functional attribute of this cytokine.
Collapse
|
28
|
Abstract
Specific allergen immunotherapy (SIT) is disease-modifying and efficacious. However, the use of whole allergen preparations is associated with frequent allergic adverse events during treatment. Many novel approaches are being designed to reduce the allergenicity of immunotherapy preparations whilst maintaining immunogenicity. One approach is the use of short synthetic peptides which representing dominant T cell epitopes of the allergen. Short peptides exhibit markedly reduced capacity to cross link IgE and activate mast cells and basophils, due to lack of tertiary structure. Murine pre-clinical studies have established the feasibility of this approach and clinical studies are currently in progress in both allergic and autoimmune diseases.
Collapse
Affiliation(s)
- Mark Larché
- Department of Allergy and Clinical Immunology, Faculty of Medicine, Imperial College, South Kensington, London
| |
Collapse
|
29
|
Van Eden W, Wick G, Albani S, Cohen I. Stress, Heat Shock Proteins, and Autoimmunity: How Immune Responses to Heat Shock Proteins Are to Be Used for the Control of Chronic Inflammatory Diseases. Ann N Y Acad Sci 2007; 1113:217-37. [PMID: 17584980 DOI: 10.1196/annals.1391.020] [Citation(s) in RCA: 105] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Especially since the (re-)discovery of T cell subpopulations with specialized regulatory activities, mechanisms of anti-inflammatory T cell regulation are studied very actively and are expected to lead to the development of novel immunotherapeutic approaches, especially in chronic inflammatory diseases. Heat shock proteins (Hsp) are possible targets for regulatory T cells due to their enhanced expression in inflamed (stressed) tissues and the evidence that Hsp induce anti-inflammatory immunoregulatory T cell responses. Initial evidence for an immunoregulatory role of Hsp in chronic inflammation was obtained through analysis of T cell responses in the rat model of adjuvant arthritis and the findings that Hsp immunizations protected against the induction of various forms of autoimmune arthritis in rat and mouse models. Since then, immune reactivity to Hsp was found to result from inflammation in various disease models and human inflammatory conditions, such as rheumatoid arthritis (RA), type 1 diabetes, and atherosclerosis. Now, also in the light of a growing interest in T cell regulation, it is of interest to further explore the mechanisms through which Hsp can be utilized to trigger immunoregulatory pathways, capable of suppressing such a wide and diversified spectrum of inflammatory diseases.
Collapse
Affiliation(s)
- Willem Van Eden
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Yalelaan 1, Utrecht University, 3584CL Utrecht, the Netherlands.
| | | | | | | |
Collapse
|
30
|
Abstract
Specific allergen immunotherapy has been widely practised for almost 100 years. Whilst this approach is disease-modifying and efficacious, the use of whole allergen preparations is associated with an unacceptably high prevalence of allergic adverse events during treatment. Many approaches to reduce the allergenicity of immunotherapy preparations whilst maintaining immunogenicity are under development. One such approach is the use of short synthetic peptides which represent major T-cell epitopes of the allergen. Major potential advantages of this approach include markedly reduced capacity to cross-link immunoglobulin-E and activate mast cells and basophils, and ease of manufacture and standardization. Promising results in preclinical studies have led to the translation of this approach to clinical studies in humans. Peptide immunotherapy is currently under development for allergic and autoimmune diseases.
Collapse
Affiliation(s)
- M Larché
- Division of Clinical Immunology & Allergy, Department of Medicine, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
31
|
Roord ST, Zonneveld-Huijssoon E, Le T, Yung GP, Koffeman E, Ronaghy A, Ghahramani N, Lanza P, Billetta R, Prakken BJ, Albani S. Modulation of T cell function by combination of epitope specific and low dose anticytokine therapy controls autoimmune arthritis. PLoS One 2006; 1:e87. [PMID: 17183718 PMCID: PMC1762388 DOI: 10.1371/journal.pone.0000087] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2006] [Accepted: 11/09/2006] [Indexed: 12/16/2022] Open
Abstract
Innate and adaptive immunity contribute to the pathogenesis of autoimmune arthritis by generating and maintaining inflammation, which leads to tissue damage. Current biological therapies target innate immunity, eminently by interfering with single pro-inflammatory cytokine pathways. This approach has shown excellent efficacy in a good proportion of patients with Rheumatoid Arthritis (RA), but is limited by cost and side effects. Adaptive immunity, particularly T cells with a regulatory function, plays a fundamental role in controlling inflammation in physiologic conditions. A growing body of evidence suggests that modulation of T cell function is impaired in autoimmunity. Restoration of such function could be of significant therapeutic value. We have recently demonstrated that epitope-specific therapy can restore modulation of T cell function in RA patients. Here, we tested the hypothesis that a combination of anti-cytokine and epitope-specific immunotherapy may facilitate the control of autoimmune inflammation by generating active T cell regulation. This novel combination of mucosal tolerization to a pathogenic T cell epitope and single low dose anti-TNFα was as therapeutically effective as full dose anti-TNFα treatment. Analysis of the underlying immunological mechanisms showed induction of T cell immune deviation.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antigens, Bacterial/genetics
- Antigens, Bacterial/immunology
- Arthritis, Experimental/immunology
- Arthritis, Experimental/pathology
- Arthritis, Experimental/therapy
- Autoimmunity
- Base Sequence
- Chaperonin 60/genetics
- Chaperonin 60/immunology
- Cytokines/antagonists & inhibitors
- Cytokines/genetics
- Cytokines/metabolism
- DNA Primers/genetics
- Epitopes, T-Lymphocyte/genetics
- Epitopes, T-Lymphocyte/immunology
- Etanercept
- Hindlimb/pathology
- Humans
- Immunity, Mucosal
- Immunoglobulin G/administration & dosage
- Immunotherapy/methods
- Immunotherapy, Adoptive
- Male
- Mycobacterium tuberculosis/genetics
- Mycobacterium tuberculosis/immunology
- Rats
- Rats, Inbred Lew
- Receptors, Tumor Necrosis Factor/administration & dosage
- T-Lymphocytes/immunology
- T-Lymphocytes, Regulatory/immunology
- Tumor Necrosis Factor-alpha/antagonists & inhibitors
Collapse
Affiliation(s)
- Sarah T.A. Roord
- Department of Medicine, University of California San DiegoLa Jolla, California, United States of America
- Department of Pediatrics, University of California San DiegoLa Jolla, California, United States of America
- Department of Pediatric Immunology, University Medical Center Utrecht, Wilhelmina Children's HospitalUtrecht, The Netherlands
- Immunology Advanced Center On Preclinical Immunogenomics project, EUREKA Institute for Translational MedicineSiracusa, Italy
| | - Evelien Zonneveld-Huijssoon
- Department of Pediatric Immunology, University Medical Center Utrecht, Wilhelmina Children's HospitalUtrecht, The Netherlands
- Immunology Advanced Center On Preclinical Immunogenomics project, EUREKA Institute for Translational MedicineSiracusa, Italy
| | - Tho Le
- Department of Medicine, University of California San DiegoLa Jolla, California, United States of America
- Department of Pediatrics, University of California San DiegoLa Jolla, California, United States of America
- Immunology Advanced Center On Preclinical Immunogenomics project, EUREKA Institute for Translational MedicineSiracusa, Italy
| | - Gisella Puga Yung
- Department of Medicine, University of California San DiegoLa Jolla, California, United States of America
- Department of Pediatrics, University of California San DiegoLa Jolla, California, United States of America
- Immunology Advanced Center On Preclinical Immunogenomics project, EUREKA Institute for Translational MedicineSiracusa, Italy
| | - Eva Koffeman
- Department of Medicine, University of California San DiegoLa Jolla, California, United States of America
- Department of Pediatrics, University of California San DiegoLa Jolla, California, United States of America
- Immunology Advanced Center On Preclinical Immunogenomics project, EUREKA Institute for Translational MedicineSiracusa, Italy
| | - Arash Ronaghy
- Department of Medicine, University of California San DiegoLa Jolla, California, United States of America
| | - Negar Ghahramani
- Androclus TherapeuticsSan Diego, California, United States of America
| | - Paola Lanza
- Androclus TherapeuticsSan Diego, California, United States of America
| | - Rosario Billetta
- Androclus TherapeuticsSan Diego, California, United States of America
| | - Berent J. Prakken
- Department of Pediatric Immunology, University Medical Center Utrecht, Wilhelmina Children's HospitalUtrecht, The Netherlands
- Immunology Advanced Center On Preclinical Immunogenomics project, EUREKA Institute for Translational MedicineSiracusa, Italy
| | - Salvatore Albani
- Department of Medicine, University of California San DiegoLa Jolla, California, United States of America
- Department of Pediatrics, University of California San DiegoLa Jolla, California, United States of America
- Androclus TherapeuticsSan Diego, California, United States of America
- Immunology Advanced Center On Preclinical Immunogenomics project, EUREKA Institute for Translational MedicineSiracusa, Italy
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
32
|
Boyle DL, Jones TL, Hammaker D, Svensson CI, Rosengren S, Albani S, Sorkin L, Firestein GS. Regulation of peripheral inflammation by spinal p38 MAP kinase in rats. PLoS Med 2006; 3:e338. [PMID: 16953659 PMCID: PMC1560929 DOI: 10.1371/journal.pmed.0030338] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2006] [Accepted: 06/12/2006] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Somatic afferent input to the spinal cord from a peripheral inflammatory site can modulate the peripheral response. However, the intracellular signaling mechanisms in the spinal cord that regulate this linkage have not been defined. Previous studies suggest spinal cord p38 mitogen-activated protein (MAP) kinase and cytokines participate in nociceptive behavior. We therefore determined whether these pathways also regulate peripheral inflammation in rat adjuvant arthritis, which is a model of rheumatoid arthritis. METHODS AND FINDINGS Selective blockade of spinal cord p38 MAP kinase by administering the p38 inhibitor SB203580 via intrathecal (IT) catheters in rats with adjuvant arthritis markedly suppressed paw swelling, inhibited synovial inflammation, and decreased radiographic evidence of joint destruction. The same dose of SB203580 delivered systemically had no effect, indicating that the effect was mediated by local concentrations in the neural compartment. Evaluation of articular gene expression by quantitative real-time PCR showed that spinal p38 inhibition markedly decreased synovial interleukin-1 and -6 and matrix metalloproteinase (MMP3) gene expression. Activation of p38 required tumor necrosis factor alpha (TNFalpha) in the nervous system because IT etanercept (a TNF inhibitor) given during adjuvant arthritis blocked spinal p38 phosphorylation and reduced clinical signs of adjuvant arthritis. CONCLUSIONS These data suggest that peripheral inflammation is sensed by the central nervous system (CNS), which subsequently activates stress-induced kinases in the spinal cord via a TNFalpha-dependent mechanism. Intracellular p38 MAP kinase signaling processes this information and profoundly modulates somatic inflammatory responses. Characterization of this mechanism could have clinical and basic research implications by supporting development of new treatments for arthritis and clarifying how the CNS regulates peripheral immune responses.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/administration & dosage
- Anti-Inflammatory Agents, Non-Steroidal/pharmacology
- Arthritis, Experimental/metabolism
- Arthritis, Experimental/pathology
- Arthritis, Experimental/prevention & control
- Arthritis, Rheumatoid/metabolism
- Arthritis, Rheumatoid/pathology
- Arthritis, Rheumatoid/prevention & control
- Cell Proliferation/drug effects
- Cells, Cultured
- Disease Models, Animal
- Imidazoles/administration & dosage
- Imidazoles/pharmacology
- Injections, Spinal
- Interleukin-1/metabolism
- Interleukin-6/metabolism
- Joints/drug effects
- Joints/pathology
- Matrix Metalloproteinase 3/metabolism
- Phosphorylation
- Protein Kinase Inhibitors/administration & dosage
- Protein Kinase Inhibitors/pharmacology
- Pyridines/administration & dosage
- Pyridines/pharmacology
- RNA, Messenger/metabolism
- Rats
- Rats, Inbred Lew
- Spinal Cord/drug effects
- Spinal Cord/enzymology
- Synovial Membrane/drug effects
- Synovial Membrane/metabolism
- Synovial Membrane/pathology
- T-Lymphocytes/drug effects
- Time Factors
- Tumor Necrosis Factor-alpha/metabolism
- p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
- p38 Mitogen-Activated Protein Kinases/metabolism
Collapse
Affiliation(s)
- David L Boyle
- Division of Rheumatology, Allergy, and Immunology, University of California San Diego School of Medicine, La Jolla, California, United States of America
| | - Toni L Jones
- Department of Anesthesiology, University of California San Diego School of Medicine, La Jolla, California, United States of America
| | - Deepa Hammaker
- Division of Rheumatology, Allergy, and Immunology, University of California San Diego School of Medicine, La Jolla, California, United States of America
| | - Camille I Svensson
- Department of Anesthesiology, University of California San Diego School of Medicine, La Jolla, California, United States of America
| | - Sanna Rosengren
- Division of Rheumatology, Allergy, and Immunology, University of California San Diego School of Medicine, La Jolla, California, United States of America
| | - Salvatore Albani
- Division of Rheumatology, Allergy, and Immunology, University of California San Diego School of Medicine, La Jolla, California, United States of America
| | - Linda Sorkin
- Department of Anesthesiology, University of California San Diego School of Medicine, La Jolla, California, United States of America
| | - Gary S Firestein
- Division of Rheumatology, Allergy, and Immunology, University of California San Diego School of Medicine, La Jolla, California, United States of America
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
33
|
Anderson PO, Manzo BA, Sundstedt A, Minaee S, Symonds A, Khalid S, Rodriguez-Cabezas ME, Nicolson K, Li S, Wraith DC, Wang P. Persistent antigenic stimulation alters the transcription program in T cells, resulting in antigen-specific tolerance. Eur J Immunol 2006; 36:1374-85. [PMID: 16708405 PMCID: PMC2652694 DOI: 10.1002/eji.200635883] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Repetitive antigen stimulation induces peripheral T cell tolerance in vivo. It is not known, however, whether multiple stimulations merely suppress T cell activation or, alternatively, change the transcriptional program to a distinct, tolerant state. In this study, we have discovered that STAT3 and STAT5 were activated in response to antigen stimulation in vivo, in marked contrast to the suppression of AP-1, NF-kappaB and NFAT. In addition, a number of transcription factors were induced in tolerant T cells following antigen challenge in vivo, including T-bet, Irf-1 and Egr-2. The altered transcription program in tolerant cells associates closely with the suppression of cell cycle progression and IL-2 production, as well as with the induction of IL-10. Studies of T-bet and Egr-2 show that the function of T-bet in peptide treatment-induced regulatory T cells is not associated with Th1 differentiation, but correlates with the suppression of IL-2, whereas expression of Egr-2 led to an up-regulation of the cell cycle inhibitors p21(cip1) and p27(kip). Our results demonstrate a balanced transcription program regulated by different transcription factors for T cell activation and/or tolerance during antigen-induced T cell responses. Persistent antigen stimulation can induce T cell tolerance by changing the balance of transcription factors.
Collapse
Affiliation(s)
- Per O. Anderson
- Institute of Cell and Molecular Science, Barts and London School of Medicine, London, UK
| | - Barbara A. Manzo
- Institute of Cell and Molecular Science, Barts and London School of Medicine, London, UK
| | - Anette Sundstedt
- Department of Pathology and Microbiology, School of Medical Sciences, University of Bristol, Bristol, UK
| | - Sophie Minaee
- Department of Pathology and Microbiology, School of Medical Sciences, University of Bristol, Bristol, UK
| | - Alistair Symonds
- Institute of Cell and Molecular Science, Barts and London School of Medicine, London, UK
| | - Sabah Khalid
- Microarray Group, Department of Biological Sciences, Brunel University, Uxbridge, London, UK
| | | | - Kirsty Nicolson
- Department of Pathology and Microbiology, School of Medical Sciences, University of Bristol, Bristol, UK
| | - Suling Li
- Microarray Group, Department of Biological Sciences, Brunel University, Uxbridge, London, UK
| | - David C. Wraith
- Department of Pathology and Microbiology, School of Medical Sciences, University of Bristol, Bristol, UK
| | - Ping Wang
- Institute of Cell and Molecular Science, Barts and London School of Medicine, London, UK
| |
Collapse
|
34
|
Root-Bernstein R, Couturier J. Antigenic complementarity in the origins of autoimmunity: a general theory illustrated with a case study of idiopathic thrombocytopenia purpura. Clin Dev Immunol 2006; 13:49-65. [PMID: 16603444 PMCID: PMC2270743 DOI: 10.1080/17402520600578731] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
We describe a novel, testable theory of autoimmunity, outline novel predictions made by the theory, and illustrate its application to unravelling the possible causes of idiopathic thrombocytopenia purpura (ITP). Pairs of stereochemically complementary antigens induce complementary immune responses (antibody or T-cell) that create loss of regulation and civil war within the immune system itself. Antibodies attack antibodies creating circulating immune complexes; T-cells attack T-cells creating perivascular cuffing. This immunological civil war abrogates the self-nonself distinction. If at least one of the complementary antigens mimics a self antigen, then this unregulated immune response will target host tissues as well. Data demonstrating that complementary antigens are found in some animal models of autoimmunity and may be present in various human diseases, especially ITP, are reviewed. Specific mechanisms for preventing autoimmunity or suppressing existing autoimmunity are derived from the theory, and critical tests proposed. Finally, we argue that Koch's postulates are inadequate for establishing disease causation for multiple-antigen diseases and discuss the possibility that current research has failed to elucidate the causes of human autoimmune diseases because we are using the wrong criteria.
Collapse
|
35
|
|
36
|
Kamphuis S, Albani S, Prakken BJ. Heat-shock protein 60 as a tool for novel therapeutic strategies that target the induction of regulatory T cells in human arthritis. Expert Opin Biol Ther 2006; 6:579-89. [PMID: 16706605 DOI: 10.1517/14712598.6.6.579] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
In health, immune responses to self are abundantly available, but under strict control of mechanisms of peripheral tolerance. Occasionally the immune system loses control and an autoimmune disease develops. At present, treatment of autoimmune disease is based on generalised suppression of all immune responses, and is often needed to be lifelong, leading to long-term toxicities and suppression of protective immune responses against pathogens. A more targeted approach would be to reset the immune system via restoration of failing regulatory mechanisms, and redirect the immune system to a state of tolerance. Over the past decade there have been enormous advances in the understanding of basic processes that control immune tolerance, pushing regulatory T cells forward as targets for novel therapeutic strategies. This review describes the development of antigen-specific immunotherapy that targets the antigen-specific induction of regulatory T cells as a means to treat autoimmune disease. The 'holy grail' for autoimmunity is not the disease-causing antigen, but the disease-curing antigen.
Collapse
Affiliation(s)
- Sylvia Kamphuis
- Department of Paediatric Immunology, IACOPO Institute for Translational Medicine, Wilhelmina Children's Hospital, University Medical Center Utrecht, The Netherlands
| | | | | |
Collapse
|
37
|
Abstract
Synthetic peptides representing T-cell epitopes of allergens and autoantigens have been employed to induce antigen-specific tolerance in vivo in experimental models and the clinical setting. Delivery of peptides orally or by injection leads to reduced reactivity to antigen accompanied by the induction of T cells with a regulatory phenotype. Peptide therapy may provide a safe, effective, and economically viable approach for disease-modifying therapy in autoimmune and allergic diseases.
Collapse
Affiliation(s)
- Mark Larché
- Department of Allergy and Clinical Immunology, Faculty of Medicine, Imperial College, South Kensington, London SW7 2AZ, UK.
| |
Collapse
|
38
|
Root-Bernstein R. A measles-derived peptide treats and vaccinates against adjuvant arthritis. Clin Exp Rheumatol 2006. [DOI: 10.1016/j.autrev.2006.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
39
|
de Kleer IM, Albani S, Prakken BJ. T-cell regulation in juvenile idiopathic arthritis. ACTA ACUST UNITED AC 2006. [DOI: 10.2217/17460816.1.1.111] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
40
|
Senger S, Maurano F, Mazzeo MF, Gaita M, Fierro O, David CS, Troncone R, Auricchio S, Siciliano RA, Rossi M. Identification of Immunodominant Epitopes of α-Gliadin in HLA-DQ8 Transgenic Mice following Oral Immunization. THE JOURNAL OF IMMUNOLOGY 2005; 175:8087-95. [PMID: 16339546 DOI: 10.4049/jimmunol.175.12.8087] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Celiac disease, triggered by wheat gliadin and related prolamins from barley and rye, is characterized by a strong association with HLA-DQ2 and HLA-DQ8 genes. Gliadin is a mixture of many proteins that makes difficult the identification of major immunodominant epitopes. To address this issue, we expressed in Escherichia coli a recombinant alpha-gliadin (r-alpha-gliadin) showing the most conserved sequence among the fraction of alpha-gliadins. HLA-DQ8 mice, on a gluten-free diet, were intragastrically immunized with a chymotryptic digest of r-alpha-gliadin along with cholera toxin as adjuvant. Spleen and mesenteric lymph node T cell responses were analyzed for in vitro proliferative assay using a panel of synthetic peptides encompassing the entire sequence of r-alpha-gliadin. Two immunodominant epitopes corresponding to peptide p13 (aa 120-139) and p23 (aa 220-239) were identified. The response was restricted to DQ and mediated by CD4+ T cells. In vitro tissue transglutaminase deamidation of both peptides did not increase the response; furthermore, tissue transglutaminase catalyzed extensive deamidation in vitro along the entire r-alpha-gliadin molecule, but failed to elicit new immunogenic determinants. Surprisingly, the analysis of the cytokine profile showed that both deamidated and native peptides induced preferentially IFN-gamma secretion, despite the use of cholera toxin, a mucosal adjuvant that normally induces a Th2 response to bystander Ags. Taken together, these data suggest that, in this model of gluten hypersensitivity, deamidation is not a prerequisite for the initiation of gluten responses.
Collapse
Affiliation(s)
- Stefania Senger
- Istituto di Scienze dell'Alimentazione, Consiglio Nazionale delle Ricerche, Avellino, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Eden WV. Immunoregulatory T-Cell Response to Heat Shock Proteins and Suppression of Experimental Autoimmunity. Transfus Med Hemother 2005. [DOI: 10.1159/000089129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
42
|
Brentano F, Kyburz D, Schorr O, Gay R, Gay S. The role of Toll-like receptor signalling in the pathogenesis of arthritis. Cell Immunol 2005; 233:90-6. [PMID: 15963480 DOI: 10.1016/j.cellimm.2005.04.018] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2005] [Accepted: 04/21/2005] [Indexed: 02/07/2023]
Abstract
Recent evidence highlighted the role of Toll-like receptors (TLRs) as key recognition structures of the innate immune system. The activation of TLRs initiates the production of inflammatory cytokines, chemokines, tissue destructive enzymes, and type I interferons. In addition, TLR signalling plays an important role in the activation and direction of the adaptive immune system by the upregulation of costimulatory molecules of antigen presenting cells. Considering the important role of TLR signalling as a critical link between innate and adaptive immunity it has been proposed that a dysregulation in TLR signalling might be associated with autoimmunity. In this review, recent studies on TLR signal transduction pathways activated by corresponding ligands are summarized and evidence for a possible role of TLR signalling in the pathogenesis of rheumatoid arthritis is discussed.
Collapse
Affiliation(s)
- Fabia Brentano
- Center of Experimental Rheumatology, University Hospital, CH-8091 Zurich, Switzerland.
| | | | | | | | | |
Collapse
|
43
|
Mia MY, Durai M, Kim HR, Moudgil KD. Heat shock protein 65-reactive T cells are involved in the pathogenesis of non-antigenic dimethyl dioctadecyl ammonium bromide-induced arthritis. THE JOURNAL OF IMMUNOLOGY 2005; 175:219-27. [PMID: 15972652 DOI: 10.4049/jimmunol.175.1.219] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Dimethyl dioctadecyl ammonium bromide (DDA) (C(38)H(80)NBr) is a nonantigenic lipoid material. DDA-induced arthritis (DIA) in the Lewis (LEW) (RT.1(l)) rat is a new experimental model for human rheumatoid arthritis (RA). DIA is a T cell-mediated autoimmune disease. However, the precise self/foreign Ags associated with the disease process in DIA are not yet known. We observed that LEW rats with DIA spontaneously raised a vigorous T cell response both to 65-kDa self (rat) heat shock protein (Rhsp65) and mycobacterial hsp65 (Bhsp65), but not to another arthritis-related Ag, bovine collagen type II. The T cell response to Rhsp65 was focused predominantly on determinant regions 120-134 and 213-227 of the self protein. Interestingly, pretreatment of adult LEW rats using either a mixture of peptides 120-134 and 213-227 of Rhsp65 or a low nonarthritogenic dose of DDA induced protection against subsequent DIA. Intriguingly, the protection induced by the latter was associated with spontaneous priming of T cells specific for peptide 213-227 of Rhsp65. Similarly, LEW rats neonatally tolerized against either Rhsp65 or Bhsp65 were significantly protected from subsequently induced DIA at adult stage, showing the disease-modulating attribute of the hsp65-specific T cells. Taken together, the above findings demonstrate that the hsp65-directed T cell repertoire is of significance in the pathogenesis of autoimmune arthritis induced by nonantigenic DDA. Like other animal models of RA involving hsp65, these first insights into the disease-associated Ags in the DIA model would pave the way for further understanding of the immunological aspects of induction and regulation of RA.
Collapse
Affiliation(s)
- Md Younus Mia
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | | | | | | |
Collapse
|
44
|
Larché M. Peptide therapy for allergic diseases: basic mechanisms and new clinical approaches. Pharmacol Ther 2005; 108:353-61. [PMID: 16014312 DOI: 10.1016/j.pharmthera.2005.05.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2005] [Accepted: 05/16/2005] [Indexed: 11/22/2022]
Abstract
Desensitising allergen immunotherapy has been practised for many decades. Although time consuming, this form of therapy is antigen-specific and disease-modifying, in contrast to palliative pharmacotherapy. However, the use of allergen extracts containing native allergen molecules frequently results in allergic adverse reactions to treatment. Several strategies to reduce the allergenicity of therapeutic preparations, while maintaining their therapeutic benefit, are being developed. Peptide immunotherapy is one such approach. Short synthetic peptides, comprising T cell epitopes of major allergens, were unable to crosslink allergen-specific IgE molecules on basophils in vitro. Treatment of allergic volunteers with allergen peptides resulted in reduced skin, lung and nasal sensitivity to allergen challenge and improved their subjective ability to tolerate allergen exposure. Peptides reduced pro-inflammatory cytokine secretion from peripheral blood cells, whilst increasing the immunosuppressive cytokine IL-10. Furthermore, peptide therapy was associated with the induction of a population of CD4+ T cells with a suppressive functional phenotype. Thus, peptide therapy may be suitable for the antigen-specific treatment of allergic diseases.
Collapse
Affiliation(s)
- Mark Larché
- Department of Allergy and Clinical Immunology, Imperial College London, Faculty of Medicine, National Heart and Lung Institute, Dovehouse Street, London SW3 6LY, United Kingdom.
| |
Collapse
|
45
|
Larché M, Wraith DC. Peptide-based therapeutic vaccines for allergic and autoimmune diseases. Nat Med 2005; 11:S69-76. [PMID: 15812493 DOI: 10.1038/nm1226] [Citation(s) in RCA: 253] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Allergic and autoimmune diseases are forms of immune hypersensitivity that increasingly cause chronic ill health. Most current therapies treat symptoms rather than addressing underlying immunological mechanisms. The ability to modify antigen-specific pathogenic responses by therapeutic vaccination offers the prospect of targeted therapy resulting in long-term clinical improvement without nonspecific immune suppression. Examples of specific immune modulation can be found in nature and in established forms of immune desensitization. Understanding and exploiting common mechanisms such as the ability to induce antigen-specific regulatory cells should allow the development of effective therapeutic strategies for both forms of immunopathology. Targeting pathogenic T cells using vaccines consisting of synthetic peptides representing T cell epitopes is one such strategy that is currently being evaluated with encouraging results. Future challenges in the development of therapeutic vaccines include selection of appropriate antigens and peptides, optimization of peptide dose and route of administration and identifying strategies to induce bystander suppression.
Collapse
Affiliation(s)
- Mark Larché
- Department of Allergy & Clinical Immunology, Imperial College London, Faculty of Medicine, Dovehouse Street, London, SW3 6LY, UK.
| | | |
Collapse
|
46
|
van Eden W, van der Zee R, Prakken B. Heat-shock proteins induce T-cell regulation of chronic inflammation. Nat Rev Immunol 2005; 5:318-30. [PMID: 15803151 DOI: 10.1038/nri1593] [Citation(s) in RCA: 393] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Immune responses to certain heat-shock proteins (HSPs) develop in almost all inflammatory diseases; however, the significance of such responses is only now becoming clear. In experimental disease models, HSPs can prevent or arrest inflammatory damage, and in initial clinical trials in patients with chronic inflammatory disease, HSP-derived peptides have been shown to promote the production of anti-inflammatory cytokines, indicating that HSPs have immunoregulatory potential. In this Review, we discuss the unique characteristics of HSPs that endow them with these immunoregulatory qualities.
Collapse
Affiliation(s)
- Willem van Eden
- Division of Immunology, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584CL Utrecht, The Netherlands.
| | | | | |
Collapse
|
47
|
Boots AM, Verhaert PD, te Poele RJ, Evers S, Coenen-de Roo CJ, Cleven J, Bos ES. Antigens up the nose: identification of putative biomarkers for nasal tolerance induction functional studies combined with proteomics. J Proteome Res 2005; 3:1056-62. [PMID: 15473695 DOI: 10.1021/pr049907c] [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: 01/03/2023]
Abstract
Intranasal autoantigen delivery is the most effective means of inducing mucosal tolerance and suppression of autoimmune disease. In an effort to identify markers of the "tolerant state", we employed proteomics technology at the level of the cervical lymph node. The analysis revealed that nasal antigen administration (without adiuvant) led to modulation of various proteins among which the most prominent were haptoglobin, nonintegrin 67 kDa laminin receptor, and MRP8. The immunoregulatory haptoglobin may qualify as (bio)marker for effective immunotherapy.
Collapse
MESH Headings
- Adipokines
- Animals
- Antigen Presentation/immunology
- Biomarkers/analysis
- Biomarkers/metabolism
- Calgranulin A/analysis
- Calgranulin A/metabolism
- Carrier Proteins/analysis
- Carrier Proteins/metabolism
- Cell Line
- Chitinase-3-Like Protein 1
- Cytokines/metabolism
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Down-Regulation/immunology
- Electrophoresis, Gel, Two-Dimensional
- Glycoproteins/immunology
- Haptoglobins/analysis
- Haptoglobins/metabolism
- Immune Tolerance/immunology
- Immunization
- Lectins
- Lymph Nodes/chemistry
- Lymph Nodes/cytology
- Lymph Nodes/immunology
- Lymphocyte Activation/immunology
- Mass Spectrometry
- Mice
- Mice, Inbred BALB C
- Mice, Knockout
- Mice, Transgenic
- Nasal Mucosa/immunology
- Ovalbumin/immunology
- Peptide Fragments/immunology
- Protein Transport/immunology
- Proteomics
- RNA-Binding Proteins
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Laminin/analysis
- Receptors, Laminin/metabolism
- Spleen/cytology
- Spleen/immunology
- T-Lymphocytes/immunology
- Up-Regulation/immunology
Collapse
Affiliation(s)
- Annemieke M Boots
- Department of Target Discovery and Pharmacology at NV Organon, Oss, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
48
|
Stemmer C, Guichard G. Antigen-based T-cell-targeted immunotherapy: recent developments in autoimmunity and allergy. Expert Opin Ther Pat 2005. [DOI: 10.1517/13543776.8.7.819] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
49
|
Abstract
Allergic diseases such as asthma, rhinitis, and eczema are increasing in prevalence and affect up to 15% of populations in Westernized countries. The description of Tregs as T cells that prevent development of autoimmune disease led to considerable interest in whether these Tregs were also normally involved in prevention of sensitization to allergens and whether it might be possible to manipulate Tregs for the therapy of allergic disease. Current data suggest that Th2 responses to allergens are normally suppressed by both CD4+CD25+ Tregs and IL-10 Tregs. Furthermore, suppression by these subsets is decreased in allergic individuals. In animal models, Tregs could be induced by high- or low-dose inhaled antigen, and prior induction of such Tregs prevented subsequent development of allergen sensitization and airway inflammation in inhaled challenge models. For many years, allergen-injection immunotherapy has been used for the therapy of allergic disease, and this treatment may induce IL-10 Tregs, leading to both suppression of Th2 responses and a switch from IgE to IgG4 antibody production. Improvements in allergen immunotherapy, such as peptide therapy, and greater understanding of the biology of Tregs hold great promise for the treatment and prevention of allergic disease.
Collapse
Affiliation(s)
- Douglas S Robinson
- Department of Allergy and Clinical Immunology, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
| | | | | |
Collapse
|
50
|
Abstract
Allergic diseases such as asthma, rhinitis, and eczema are increasing in prevalence and affect up to 15% of populations in Westernized countries. The description of Tregs as T cells that prevent development of autoimmune disease led to considerable interest in whether these Tregs were also normally involved in prevention of sensitization to allergens and whether it might be possible to manipulate Tregs for the therapy of allergic disease. Current data suggest that Th2 responses to allergens are normally suppressed by both CD4+CD25+ Tregs and IL-10 Tregs. Furthermore, suppression by these subsets is decreased in allergic individuals. In animal models, Tregs could be induced by high- or low-dose inhaled antigen, and prior induction of such Tregs prevented subsequent development of allergen sensitization and airway inflammation in inhaled challenge models. For many years, allergen-injection immunotherapy has been used for the therapy of allergic disease, and this treatment may induce IL-10 Tregs, leading to both suppression of Th2 responses and a switch from IgE to IgG4 antibody production. Improvements in allergen immunotherapy, such as peptide therapy, and greater understanding of the biology of Tregs hold great promise for the treatment and prevention of allergic disease.
Collapse
Affiliation(s)
- Douglas S Robinson
- Department of Allergy and Clinical Immunology, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
| | | | | |
Collapse
|