1
|
Wu L, Zhao K, Xu L, Cui J, Ruan L, Bei S, Cao J, Qi X, Shen S. Macrophages-mediated delivery of protoporphyrin for sonodynamic therapy of rheumatoid arthritis. ULTRASONICS SONOCHEMISTRY 2024; 107:106928. [PMID: 38820932 PMCID: PMC11179255 DOI: 10.1016/j.ultsonch.2024.106928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 05/21/2024] [Accepted: 05/25/2024] [Indexed: 06/02/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic systemic inflammatory disease characterized by infiltration of inflammatory cells, hyperplasia of synovium, and destruction of the joint cartilage. Owing to the low drug delivery efficiency and limited immunosuppression effect, complete cure for RA remains a formidable challenge. Here, we show that live macrophages (Mφs) carrying protoporphyrin-loaded Fe3O4 nanoparticles can migrate to the RA tissues and inhibit the inflammation by sonodynamic therapy. The inflammation of RA leads to the release of cytokines, which guides the migration of the Mφs into the RA tissues, realizing precise delivery of therapeutics. The following sonodynamic therapy induced by ultrasound and protoporphyrin destructs the proliferating synovial cells and also infiltrated inflammatory cells, demonstrating significant therapeutic effect for RA. Meanwhile, the cytokines and relapse of RA can be remarkably suppressed because of the efficient damage to the resident inflammatory cells.
Collapse
Affiliation(s)
- Lin Wu
- Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Kai Zhao
- Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China; College of Pharmaceutical Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Leyuan Xu
- College of Pharmaceutical Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Junming Cui
- Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China; College of Pharmaceutical Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Li Ruan
- Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China; College of Pharmaceutical Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Shifang Bei
- Affiliated People's Hospital of Jiangsu University, Zhenjiang 212002, China.
| | - Jin Cao
- College of Pharmaceutical Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Xueyong Qi
- College of Pharmaceutical Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Song Shen
- College of Pharmaceutical Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China.
| |
Collapse
|
2
|
Zhao Y, Chen X, He P, Wang X, Xu Y, Hu R, Ou Y, Zhang Z, Zhang Z, Du G, Sun X. Transdermal Microneedles Alleviated Rheumatoid Arthritis by Inducing Immune Tolerance via Skin-Resident Antigen Presenting Cells. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2307366. [PMID: 38039446 DOI: 10.1002/smll.202307366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 10/26/2023] [Indexed: 12/03/2023]
Abstract
Restoring immune tolerance is the ultimate goal for rheumatoid arthritis (RA) treatment. The most reported oral or intravenous injection routes for the immunization of autoantigens cause gastrointestinal side effects, low patient compliance, and unsatisfied immune tolerance induction. Herein, the use of a transdermal microneedle patch is for the first time investigated to codeliver CII peptide autoantigen and rapamycin for reversing immune disorders of RA. The immunized microneedles efficiently recruit antigen-presenting cells particularly Langerhans cells, and induce tolerogenic dendritic cells at the administration skin site. The tolerogenic dendritic cells further homing to lymph nodes to activate systemic Treg cell differentiation, which upregulates the expression of anti-inflammatory mediators while inhibiting the polarization of Th1/2 and Th17 T cell phenotypes and the expression of inflammatory profiles. As a result, the optimized microneedles nearly completely eliminate RA symptoms and inflammatory infiltrations. Furthermore, it is demonstrated that a low dose of rapamycin is crucial for the successful induction of immune tolerance. The results indicate that a rationally designed microneedle patch is a promising strategy for immune balance restoration with increased immune tolerance induction efficiency and patient compliance.
Collapse
Affiliation(s)
- Yuanhao Zhao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Xiaoyan Chen
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Penghui He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Xuanyu Wang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Yanhua Xu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Rui Hu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Yangsen Ou
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Zhihua Zhang
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Zhibing Zhang
- School of Chemical Engineering, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Guangsheng Du
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Xun Sun
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
3
|
From vaccines to nanovaccines: A promising strategy to revolutionize rheumatoid arthritis treatment. J Control Release 2022; 350:107-121. [PMID: 35977582 DOI: 10.1016/j.jconrel.2022.08.020] [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: 07/06/2022] [Revised: 08/08/2022] [Accepted: 08/10/2022] [Indexed: 10/15/2022]
Abstract
Rheumatoid arthritis (RA) is a joint-related autoimmune disease that is difficult to cure. Most therapeutics act to alleviate the symptoms but not correct the causes of RA. Novel strategies that specifically target the causes are highly needed for RA management. Currently, early interruption of RA is increasingly suggested but the corresponding therapeutics are not available. Vaccines that have shown great success to combat infection, cancer, degenerative diseases, autoimmune diseases, etc. are ideal candidates for a new generation of anti-RA therapeutics to correct the causes and prevent RA or interrupt RA in early phases. Anti-RA vaccines can be divided into two major categories. One is to induce neutralizing antibodies and the other is to induce antigen-specific immune tolerance. The vaccines are inherently linked to nanotechnology because they usually need a biomacromolecule or carrier to provoke sufficient immune responses. In the past decade, designed nanocarriers such as nanoparticles, liposomes, nanoemulsion, etc., have been applied to optimize the vaccines for autoimmune disease treatment. Nanotechnology endows vaccines with a higher biostability, tunable in vivo behavior, better targeting, co-delivery with stimulatory agents, regulatory effects on immune responses, etc. In this review, unmet medical needs for RA treatment and anti-RA vaccinology are first introduced. The development of anti-RA therapies from vaccines to nanovaccines are then reviewed and perspectives on how nanotechnology promotes vaccine development and advancement are finally provided. In addition, challenges for anti-RA vaccine development are summarized and advantages of nanovaccines are analyzed. In conclusion, nanovaccines will be a promising strategy to revolutionize the treatment of RA by correcting the causes in an early phase of RA.
Collapse
|
4
|
Intra-Articular Drug Delivery for Osteoarthritis Treatment. Pharmaceutics 2021; 13:pharmaceutics13122166. [PMID: 34959445 PMCID: PMC8703898 DOI: 10.3390/pharmaceutics13122166] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/13/2021] [Accepted: 12/13/2021] [Indexed: 02/07/2023] Open
Abstract
Osteoarthritis (OA) is the most prevalent degenerative joint disease affecting millions of people worldwide. Currently, clinical nonsurgical treatments of OA are only limited to pain relief, anti-inflammation, and viscosupplementation. Developing disease-modifying OA drugs (DMOADs) is highly demanded for the efficient treatment of OA. As OA is a local disease, intra-articular (IA) injection directly delivers drugs to synovial joints, resulting in high-concentration drugs in the joint and reduced side effects, accompanied with traditional oral or topical administrations. However, the injected drugs are rapidly cleaved. By properly designing the drug delivery systems, prolonged retention time and targeting could be obtained. In this review, we summarize the drugs investigated for OA treatment and recent advances in the IA drug delivery systems, including micro- and nano-particles, liposomes, and hydrogels, hoping to provide some information for designing the IA injected formulations.
Collapse
|
5
|
Meehan GR, Thomas R, Al Khabouri S, Wehr P, Hilkens CM, Wraith DC, Sieghart D, Bonelli M, Nagy G, Garside P, Tough DF, Lewis HD, Brewer JM. Preclinical models of arthritis for studying immunotherapy and immune tolerance. Ann Rheum Dis 2021; 80:1268-1277. [PMID: 34380700 PMCID: PMC8458054 DOI: 10.1136/annrheumdis-2021-220043] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 06/27/2021] [Indexed: 02/06/2023]
Abstract
Increasingly earlier identification of individuals at high risk of rheumatoid arthritis (RA) (eg, with autoantibodies and mild symptoms) improves the feasibility of preventing or curing disease. The use of antigen-specific immunotherapies to reinstate immunological self-tolerance represent a highly attractive strategy due to their potential to induce disease resolution, in contrast to existing approaches that require long-term treatment of underlying symptoms. Preclinical animal models have been used to understand disease mechanisms and to evaluate novel immunotherapeutic approaches. However, models are required to understand critical processes supporting disease development such as the breach of self-tolerance that triggers autoimmunity and the progression from asymptomatic autoimmunity to joint pain and bone loss. These models would also be useful in evaluating the response to treatment in the pre-RA period. This review proposes that focusing on immune processes contributing to initial disease induction rather than end-stage pathological consequences is essential to allow development and evaluation of novel immunotherapies for early intervention. We will describe and critique existing models in arthritis and the broader field of autoimmunity that may fulfil these criteria. We will also identify key gaps in our ability to study these processes in animal models, to highlight where further research should be targeted.
Collapse
Affiliation(s)
- Gavin R Meehan
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Ranjeny Thomas
- University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Shaima Al Khabouri
- Division of Rheumatology, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Pascale Wehr
- University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Catharien Mu Hilkens
- Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - David C Wraith
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Daniela Sieghart
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Michael Bonelli
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - György Nagy
- Department of Rheumatology & Clinical Immunology, Semmelweis University, Budapest, Hungary.,Department of Genetics, Cell and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Paul Garside
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - David F Tough
- GlaxoSmithKline Research and Development, Stevenage, Hertfordshire, UK
| | - Huw D Lewis
- GlaxoSmithKline Research and Development, Stevenage, Hertfordshire, UK
| | - James M Brewer
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| |
Collapse
|
6
|
Li C, Chen X, Luo X, Wang H, Zhu Y, Du G, Chen W, Chen Z, Hao X, Zhang Z, Sun X. Nanoemulsions Target to Ectopic Lymphoids in Inflamed Joints to Restore Immune Tolerance in Rheumatoid Arthritis. NANO LETTERS 2021; 21:2551-2561. [PMID: 33687217 DOI: 10.1021/acs.nanolett.0c05110] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Inducing immune tolerance through repeated administration of self-antigens is a promising strategy for treating rheumatoid arthritis (RA), and current research indicates that coadministration of immunomodulators can further orchestrate the tolerogenic response. However, most of the clinical trials based on tolerance induction have negligible therapeutic effects. Peripheral lymphoid organs play critical roles in immunotherapy. Here, we design an engineered nanoemulsion for targeted codelivery of self-antigens and an immunomodulator to ectopic lymphoid structures (ELSs) in inflamed joints of RA. Namely, a citrullinated multiepitope self-antigen (CitP) and rapamycin are incorporated into the nanoemulsions (NEs@CitP/Rapa), which are fabricated by a facial method using commercialized pharmaceutical excipients. After intravenous administration, the nanoemulsion shows satisfactory accumulation in the inflamed paws and provides enhanced anti-inflammatory effect in various experimental murine models of RA. Our study provides a promising targeting strategy to induce immune tolerance for the treatment of RA.
Collapse
Affiliation(s)
- Chenglong Li
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, P.R. China
| | - Xiaoyan Chen
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, P.R. China
| | - Xianjin Luo
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, P.R. China
| | - Hairui Wang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, P.R. China
| | - Yining Zhu
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, P.R. China
| | - Guangshen Du
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, P.R. China
| | - Wenfei Chen
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, P.R. China
| | - Zhengjun Chen
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, P.R. China
| | - Xinyan Hao
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, P.R. China
| | - Zhirong Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, P.R. China
| | - Xun Sun
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu 610041, P.R. China
| |
Collapse
|
7
|
Toes RE, Raza K. The autoimmune response as a potential target for tolerance induction before the development of rheumatoid arthritis. THE LANCET. RHEUMATOLOGY 2021; 3:e214-e223. [PMID: 38279384 DOI: 10.1016/s2665-9913(20)30445-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 12/07/2020] [Accepted: 12/09/2020] [Indexed: 01/28/2024]
Abstract
Rheumatoid arthritis is a chronic inflammatory disease that affects the synovial joints. Although treatment options and efficacy have increased substantially in the past two decades, the disease cannot be cured or prevented. Therefore, rheumatoid arthritis still has a considerable effect on the quality of life of patients, not only because life-long medication is often required, but also because residual disease activity leads to progressive loss of function in the musculoskeletal system and extra-articular morbidity. Key future goals in the management of rheumatoid arthritis are the ability to induce long-lasting drug-free remission in patients with the disease (ie, to achieve a cure), and to prevent disease before it emerges. To reach these goals, it is pivotal to understand the autoimmune response underlying rheumatoid arthritis pathogenesis and to develop ways to permanently silence it (ie, to induce tolerance). For preventive studies, the identification of markers (clinical, immunological, and biological) predictive of future disease is crucial, as prevention of disease will not be feasible without the ability to identify relevant at-risk target populations. In this Series paper, we review the autoimmune response underlying rheumatoid arthritis, how rheumatoid arthritis-specific autoimmunity develops and evolves during the transition from health to disease, and how tolerance studies could be designed to achieve prevention or cure of the disease.
Collapse
Affiliation(s)
- Rene Em Toes
- Department of Rheumatology, Leiden University Medical Center, Leiden, Netherlands.
| | - Karim Raza
- Research into Inflammatory Arthritis Centre Versus Arthritis and MRC-Versus Arthritis Centre for Musculoskeletal Ageing Research, Institute of Inflammation and Ageing, University of Birmingham, Queen Elizabeth Hospital, Birmingham, UK; Department of Rheumatology, Sandwell and West Birmingham NHS Trust, Birmingham, UK
| |
Collapse
|
8
|
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: 1.0] [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
|
9
|
Kumar P, Yao LJ, Saidin S, Paleja B, van Loosdregt J, Chua C, Arkachaisri T, Consolaro A, Gattorno M, Martini A, Pischel KD, Williams GW, Lotz M, Albani S. Molecular mechanisms of autophagic memory in pathogenic T cells in human arthritis. J Autoimmun 2018; 94:90-98. [PMID: 30077426 DOI: 10.1016/j.jaut.2018.07.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 07/18/2018] [Accepted: 07/18/2018] [Indexed: 01/11/2023]
Abstract
T-cell resilience is critical to the immune pathogenesis of human autoimmune arthritis. Autophagy is essential for memory T cell generation and associated with pathogenesis in rheumatoid arthritis (RA). Our aim here was to delineate the role and molecular mechanism of autophagy in resilience and persistence of pathogenic T cells from autoimmune arthritis. We demonstrated "Autophagic memory" as elevated autophagy levels in CD4+ memory T cells compared to CD4+ naive T cells and in Jurkat Human T cell line trained with starvation stress. We then showed increased levels of autophagy in pathogenic CD4+ T cells subsets from autoimmune arthritis patients. Using RNA-sequencing, transcription factor gene regulatory network and methylation analyses we identified MYC as a key regulator of autophagic memory. We validated MYC levels using qPCR and further demonstrated that inhibiting MYC increased autophagy. The present study proposes the novel concept of autophagic memory and suggests that autophagic memory confers metabolic advantage to pathogenic T cells from arthritis and supports its resilience and long term survival. Particularly, suppression of MYC imparted the heightened autophagy levels in pathogenic T cells. These studies have a direct translational valency as they identify autophagy and its metabolic controllers as a novel therapeutic target.
Collapse
Affiliation(s)
- Pavanish Kumar
- Translational Immunology Institute, SingHealth/Duke-NUS Academic Medical Centre, Singapore
| | - Leong Jing Yao
- Translational Immunology Institute, SingHealth/Duke-NUS Academic Medical Centre, Singapore
| | - Suzan Saidin
- Translational Immunology Institute, SingHealth/Duke-NUS Academic Medical Centre, Singapore
| | - Bhairav Paleja
- Translational Immunology Institute, SingHealth/Duke-NUS Academic Medical Centre, Singapore
| | - Jorg van Loosdregt
- Translational Immunology Institute, SingHealth/Duke-NUS Academic Medical Centre, Singapore
| | - Camillus Chua
- Translational Immunology Institute, SingHealth/Duke-NUS Academic Medical Centre, Singapore
| | - Thaschawee Arkachaisri
- Duke-NUS Medical School and Rheumatology and Immunology Service, KK Women's and Children's Hospital, Singapore, Singapore
| | - Alessandro Consolaro
- Second Pediatrics Division, University of Genoa and Gaslini Institute, Genova, Italy
| | - Marco Gattorno
- Second Pediatrics Division, University of Genoa and Gaslini Institute, Genova, Italy
| | - Alberto Martini
- Second Pediatrics Division, University of Genoa and Gaslini Institute, Genova, Italy
| | | | | | - Martin Lotz
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Salvatore Albani
- Translational Immunology Institute, SingHealth/Duke-NUS Academic Medical Centre, Singapore.
| |
Collapse
|
10
|
Barbera Betancourt A, Lyu Q, Broere F, Sijts A, Rutten VPMG, van Eden W. T Cell-Mediated Chronic Inflammatory Diseases Are Candidates for Therapeutic Tolerance Induction with Heat Shock Proteins. Front Immunol 2017; 8:1408. [PMID: 29123529 PMCID: PMC5662553 DOI: 10.3389/fimmu.2017.01408] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 10/11/2017] [Indexed: 12/21/2022] Open
Abstract
Failing immunological tolerance for critical self-antigens is the problem underlying most chronic inflammatory diseases of humans. Despite the success of novel immunosuppressive biological drugs, the so-called biologics, in the treatment of diseases such rheumatoid arthritis (RA) and type 1 diabetes, none of these approaches does lead to a permanent state of medicine free disease remission. Therefore, there is a need for therapies that restore physiological mechanisms of self-tolerance. Heat shock proteins (HSPs) have shown disease suppressive activities in many models of experimental autoimmune diseases through the induction of regulatory T cells (Tregs). Also in first clinical trials with HSP-based peptides in RA and diabetes, the induction of Tregs was noted. Due to their exceptionally high degree of evolutionary conservation, HSP protein sequences (peptides) are shared between the microbiota-associated bacterial species and the self-HSP in the tissues. Therefore, Treg mechanisms, such as those induced and maintained by gut mucosal tolerance for the microbiota, can play a role by targeting the more conserved HSP peptide sequences in the inflamed tissues. In addition, the stress upregulated presence of HSP in these tissues may well assist the targeting of the HSP induced Treg specifically to the sites of inflammation.
Collapse
Affiliation(s)
- Ariana Barbera Betancourt
- Faculty of Veterinary Medicine, Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, Netherlands
| | - Qingkang Lyu
- Faculty of Veterinary Medicine, Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, Netherlands
| | - Femke Broere
- Faculty of Veterinary Medicine, Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, Netherlands
| | - Alice Sijts
- Faculty of Veterinary Medicine, Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, Netherlands
| | - Victor P M G Rutten
- Faculty of Veterinary Medicine, Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, Netherlands
| | - Willem van Eden
- Faculty of Veterinary Medicine, Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
11
|
Cabrales-Rico A, Ramos Y, Besada V, del Carmen Domínguez M, Lorenzo N, García O, Alexis J, Prada D, Reyes Y, López AM, Masforrol Y, Garay H, González LJ. Development and validation of a bioanalytical method based on LC–MS/MS analysis for the quantitation of CIGB-814 peptide in plasma from Rheumatoid Arthritis patients. J Pharm Biomed Anal 2017; 143:130-140. [DOI: 10.1016/j.jpba.2017.05.030] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Revised: 05/06/2017] [Accepted: 05/14/2017] [Indexed: 11/26/2022]
|
12
|
Villar LM, Costa-Frossard L, Masterman T, Fernandez O, Montalban X, Casanova B, Izquierdo G, Coret F, Tumani H, Saiz A, Arroyo R, Fink K, Leyva L, Espejo C, Simó-Castelló M, García-Sánchez MI, Lauda F, Llufriú S, Álvarez-Lafuente R, Olascoaga J, Prada A, Oterino A, de Andrés C, Tintoré M, Ramió-Torrentà L, Rodríguez-Martín E, Picón C, Comabella M, Quintana E, Agüera E, Díaz S, Fernandez-Bolaños R, García-Merino JA, Landete L, Menéndez-González M, Navarro L, Pérez D, Sánchez-López F, Serrano-Castro PJ, Tuñón A, Espiño M, Muriel A, Bar-Or A, Álvarez-Cermeño JC. Lipid-specific immunoglobulin M bands in cerebrospinal fluid are associated with a reduced risk of developing progressive multifocal leukoencephalopathy during treatment with natalizumab. Ann Neurol 2015; 77:447-57. [PMID: 25581547 DOI: 10.1002/ana.24345] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 12/16/2014] [Accepted: 12/24/2014] [Indexed: 02/05/2023]
Affiliation(s)
- Luisa M. Villar
- Department of Immunology; Ramón y Cajal University Hospital, Institute Ramon y Cajal for Biomedical Research; Madrid Spain
- Spanish Network for the Research in Multiple Sclerosis; Spain
| | - Lucienne Costa-Frossard
- Spanish Network for the Research in Multiple Sclerosis; Spain
- Department of Neurology; Ramon y Cajal University Hospital; Institute Ramon y Cajal for Biomedical Research; Madrid Spain
| | - Thomas Masterman
- Department of Clinical Neuroscience; Karolinska Institute; Karolinska University Hospital; Huddinge Sweden
| | - Oscar Fernandez
- Spanish Network for the Research in Multiple Sclerosis; Spain
- Department of Neurology; Regional University Hospital of Malaga; Malaga Spain
| | - Xavier Montalban
- Spanish Network for the Research in Multiple Sclerosis; Spain
- Department of Neurology-Neuroimmunology; Catalunya Center for Multiple Sclerosis; Vall d'Hebron Institute of Research; Vall d'Hebron University Hospital; Autonomous University of Barcelona; Barcelona Spain
| | - Bonaventura Casanova
- Spanish Network for the Research in Multiple Sclerosis; Spain
- Department of Neurology; La Fe University Hospital; Valencia Spain
| | - Guillermo Izquierdo
- Spanish Network for the Research in Multiple Sclerosis; Spain
- Department of Neurology; Virgen Macarena University Hospital; Seville Spain
| | - Francisco Coret
- Spanish Network for the Research in Multiple Sclerosis; Spain
- Department of Neurology; Clinic University Hospital of Valencia; Valencia Spain
| | | | - Albert Saiz
- Spanish Network for the Research in Multiple Sclerosis; Spain
- Department of Neurology; Clinic Hospital and Institute August Pi i Sunyer for Biomedical Research of; University of Barcelona; Barcelona Spain
| | - Rafael Arroyo
- Spanish Network for the Research in Multiple Sclerosis; Spain
- Department of Neurology; San Carlos Clinic University Hospital; Institute for Biomedical Research of San Carlos Clinic University Hospital; Madrid Spain
| | - Katharina Fink
- Department of Clinical Neuroscience; Karolinska Institute; Karolinska University Hospital; Huddinge Sweden
| | - Laura Leyva
- Spanish Network for the Research in Multiple Sclerosis; Spain
- Department of Neurology; Regional University Hospital of Malaga; Malaga Spain
| | - Carmen Espejo
- Spanish Network for the Research in Multiple Sclerosis; Spain
- Department of Neurology-Neuroimmunology; Catalunya Center for Multiple Sclerosis; Vall d'Hebron Institute of Research; Vall d'Hebron University Hospital; Autonomous University of Barcelona; Barcelona Spain
| | - María Simó-Castelló
- Spanish Network for the Research in Multiple Sclerosis; Spain
- Department of Neurology; La Fe University Hospital; Valencia Spain
| | - María I. García-Sánchez
- Spanish Network for the Research in Multiple Sclerosis; Spain
- Department of Neurology; Virgen Macarena University Hospital; Seville Spain
| | - Florian Lauda
- Department of Neurology; University of Ulm; Ulm Germany
| | - Sara Llufriú
- Spanish Network for the Research in Multiple Sclerosis; Spain
- Department of Neurology; Clinic Hospital and Institute August Pi i Sunyer for Biomedical Research of; University of Barcelona; Barcelona Spain
| | - Roberto Álvarez-Lafuente
- Spanish Network for the Research in Multiple Sclerosis; Spain
- Department of Neurology; San Carlos Clinic University Hospital; Institute for Biomedical Research of San Carlos Clinic University Hospital; Madrid Spain
| | - Javier Olascoaga
- Spanish Network for the Research in Multiple Sclerosis; Spain
- Department of Neurology; Donostia University Hospital; San Sebastián Spain
| | - Alvaro Prada
- Spanish Network for the Research in Multiple Sclerosis; Spain
- Department of Immunology; Donostia University Hospital; San Sebastián Spain
| | - Agustín Oterino
- Department of Neurology; Marqués de Valdecilla University Hospital; Santander Spain
| | - Clara de Andrés
- Spanish Network for the Research in Multiple Sclerosis; Spain
- Department of Neurology; Gregorio Marañón University Hospital; Madrid Spain
| | - Mar Tintoré
- Spanish Network for the Research in Multiple Sclerosis; Spain
- Department of Neurology-Neuroimmunology; Catalunya Center for Multiple Sclerosis; Vall d'Hebron Institute of Research; Vall d'Hebron University Hospital; Autonomous University of Barcelona; Barcelona Spain
| | - Lluis Ramió-Torrentà
- Spanish Network for the Research in Multiple Sclerosis; Spain
- Department of Neurology; Doctor Josep Trueta University Hospital; Institute for Biomedical Research of Girona; Girona Spain
| | - Eulalia Rodríguez-Martín
- Department of Immunology; Ramón y Cajal University Hospital, Institute Ramon y Cajal for Biomedical Research; Madrid Spain
- Spanish Network for the Research in Multiple Sclerosis; Spain
| | - Carmen Picón
- Department of Immunology; Ramón y Cajal University Hospital, Institute Ramon y Cajal for Biomedical Research; Madrid Spain
- Spanish Network for the Research in Multiple Sclerosis; Spain
| | - Manuel Comabella
- Spanish Network for the Research in Multiple Sclerosis; Spain
- Department of Neurology-Neuroimmunology; Catalunya Center for Multiple Sclerosis; Vall d'Hebron Institute of Research; Vall d'Hebron University Hospital; Autonomous University of Barcelona; Barcelona Spain
| | - Ester Quintana
- Department of Neurology; Doctor Josep Trueta University Hospital; Institute for Biomedical Research of Girona; Girona Spain
| | - Eduardo Agüera
- Institute Maimónides for Biomedical Research, Reina Sofía University Hospital, University of Córdoba; Córdoba Spain
| | - Santiago Díaz
- Department of Neurology; Doctor Negrin University Hospital, Las Palmas de Gran Canaria; Spain
| | | | - Juan A. García-Merino
- Spanish Network for the Research in Multiple Sclerosis; Spain
- Department of Neurology; Puerta de Hierro University Hospital; Madrid Spain
| | - Lamberto Landete
- Department of Neurology; Doctor Peset University Hospital; Valencia Spain
| | | | - Laura Navarro
- Department of Neurology; Elche General Hospital; Elche Spain
| | - Domingo Pérez
- Department of Neurology; Hospital of El Bierzo, Ponferrada; Ponferrada Spain
| | - Fernando Sánchez-López
- Institute Maimónides for Biomedical Research, Reina Sofía University Hospital, University of Córdoba; Córdoba Spain
| | | | - Alberto Tuñón
- Department of Neurology; Central University Hospital of Asturias; Oviedo Spain
| | - Mercedes Espiño
- Department of Immunology; Ramón y Cajal University Hospital, Institute Ramon y Cajal for Biomedical Research; Madrid Spain
- Spanish Network for the Research in Multiple Sclerosis; Spain
| | - Alfonso Muriel
- Biostatistics Unit; Ramón y Cajal University Hospital; Institute Ramon y Cajal for Biomedical Research; CIBERESP; Madrid Spain
| | - Amit Bar-Or
- Montreal Neurological Institute and Hospital; McGill University; Montreal Quebec Canada
| | - José C. Álvarez-Cermeño
- Spanish Network for the Research in Multiple Sclerosis; Spain
- Department of Neurology; Ramon y Cajal University Hospital; Institute Ramon y Cajal for Biomedical Research; Madrid Spain
| |
Collapse
|
13
|
Marino LV, Pathan N, Meyer R, Wright VJ, Habibi P. The effect of 2 mMol glutamine supplementation on HSP70 and TNF-α release by LPS stimulated blood from healthy children. Clin Nutr 2014; 34:1195-201. [PMID: 25556350 DOI: 10.1016/j.clnu.2014.12.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 12/14/2014] [Accepted: 12/16/2014] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Glutamine has been shown to promote heat shock protein 70 (HSP70) release both within experimental in vitro models of sepsis (2-10 mM) and in adults post trauma (0.5 g/kg), although the efficacy varies and is dependent on the model used. The effect of glutamine supplementation on HSP70 release in children is less clear. Therefore, the aim of this study was to investigate the effect of 2 mM glutamine added to incubation media on HSP70 and inflammatory mediator release in an in vitro model of paediatric sepsis using whole blood from healthy paediatric volunteers. METHODS An in vitro whole blood endotoxin stimulation model using 1 μg/ml lipopolysaccharide (LPS) over a 24 h time period was used to investigate the effects of 2 mM glutamine on HSP70 and inflammatory mediator release in healthy children. RESULTS The addition of 2 mM glutamine to the incubation media significantly increased HSP70 release over time (p < 0.05). This was associated with an early pro-inflammatory effect on TNF-α release at 4 h (p < 0.005) which was not seen at 24 h. There was a non significant trend towards higher levels of IL-6 and IL-10 following the addition of 2 mM glutamine, which appears to differ from the response reported in adult and animal models. CONCLUSION Glutamine supplementation of incubation media promotes HSP70 and early TNF- α release in an in vitro model using blood samples from healthy children.
Collapse
Affiliation(s)
- L V Marino
- Department of Paediatrics, Imperial College, London, UK.
| | - N Pathan
- Department of Paediatrics, School of Clinical Medicine, Cambridge University, UK
| | - R Meyer
- Department of Gastroenterology, Great Ormond Street Hospital for Sick Children, London, UK
| | - V J Wright
- Department of Paediatrics, Imperial College, London, UK
| | - P Habibi
- Department of Paediatrics, Imperial College, London, UK
| |
Collapse
|
14
|
Abstract
Juvenile idiopathic arthritis (JIA)-associated uveitis is an intriguing manifestation of JIA and an important contributor of long-term damage. Its pathophysiology is still poorly understood. This review summarizes current concepts. JIA is both a multifactorial and polygenetic disease. In the past 2 decades, multiple studies have indicated that the genetic contribution to both JIA and JIA-associated uveitis is modest. From an ophthalmological point of view, much of the pathophysiological data is derived from studies in experimental uveitis animal models. The pathophysiology of the arthritic manifestations of JIA has been studied extensively in humans. These studies have focused on the principal cells of the adaptive immune system, in particular different subsets of regulatory and effector T cells, as well as on antigen presenting cells or dendritic cells. With advancing technology and advancing knowledge of the underlying mechanisms of JIA-associated uveitis, new targets for therapy might evolve in the coming years.
Collapse
Affiliation(s)
- Sebastiaan J Vastert
- Department of Pediatric Rheumatology, Wilhelmina Children's Hospital, University Medical Center Utrecht , The Netherlands and
| | | | | |
Collapse
|
15
|
Abstract
Rheumatoid arthritis (RA), the most common autoimmune disorder associated with dry eye syndrome, is also associated with sight-threatening ocular diseases such as peripheral ulcerative keratitis, scleritis and corneal melts. Tissue damage on the ocular surface of patients with RA is autoimmune-mediated. Findings from patients with dry eye have implicated defects in innate immunity (Toll-like receptors, S100A and resident antigen-presenting cells), cytokines, chemokines and T helper (TH)-cell subsets (including TH1 and TH17) in disease pathogenesis. Some of these features are probably important in dry eye related to RA, which can occur at a different time from articular disease and is more clinically severe than idiopathic dry eye. Ocular surface immune factors can be influenced by the systemic immune landscape. Depending on the severity of ocular inflammation in RA, treatment can include ciclosporin, topical corticosteroids, tacrolimus, autologous serum and systemic immunosuppression. Tissue damage is treated by inhibiting matrix metalloproteinases. Potential therapeutic strategies benefit from an improved understanding of ocular surface immunology, and include targeting of T-cell subsets, B-cell signalling or cytokines.
Collapse
|
16
|
PReS13-SPK-1587: New biologic targets for the future and basic science behind them. Pediatr Rheumatol Online J 2013. [PMCID: PMC4044788 DOI: 10.1186/1546-0096-11-s2-i17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
17
|
The effect of autoimmune arthritis treatment strategies on regulatory T-cell dynamics. Curr Opin Rheumatol 2013; 25:260-7. [PMID: 23274520 DOI: 10.1097/bor.0b013e32835d0ee4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE OF REVIEW Since their discovery over 15 years ago, intensive research has focused on the presence, phenotype and function of FOXP3(+) regulatory T cells (Treg) in autoimmune diseases such as rheumatoid arthritis (RA). The questions of whether Treg deficiencies underlie autoimmune pathology and whether or how Treg-related therapeutic approaches might be successful are still a subject of a vivid debate. In this review we give an overview of how current therapies influence Treg numbers and function in RA and juvenile idiopathic arthritis (JIA) and discuss these findings in the light of new Treg-based intervention strategies for autoimmune arthritis. RECENT FINDINGS The attempt to relate rheumatic diseases like rheumatoid arthritis and juvenile idiopathic arthritis to Treg has led to somewhat heterogeneous observations. So far, no clear defects in Treg numbers or function have been identified in autoimmune arthritis. The current standard therapies, that is methotrexate and biologicals, are generally effective, but the exact mechanism of action and their effect on Treg is not fully known. Nevertheless, the majority of in-vitro and ex-vivo data point towards a positive influence of these treatments on Treg number and function. These observations are not all consistent, however, and it is not known whether the observed effects on Treg are primary or secondary effects. To safely conduct targeted regulatory T-cell therapy in rheumatic diseases more knowledge about regulatory T-cell function in an inflammatory environment is needed that coincides with the initiative to elucidate the exact mechanism of current therapies.
Collapse
|
18
|
Arvonen M, Vähäsalo P, Turunen S, Salo HM, Mäki M, Laurila K, Vaarala O, Karttunen TJ. Altered expression of intestinal human leucocyte antigen D-related and immune signalling molecules in juvenile idiopathic arthritis. Clin Exp Immunol 2013; 170:266-73. [PMID: 23121667 DOI: 10.1111/j.1365-2249.2012.04663.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
We aimed to study intestinal immune activation status in juvenile idiopathic arthritis (JIA) by assessing intestinal human leucocyte antigen (HLA) class II expression and the mRNA expression levels of the pro- and anti-inflammatory mediators and pattern recognition receptors. HLA-D-related (HLA-DR) expression was assessed using immunohistochemical staining of frozen sections in 11 children with JIA and 17 controls. The gene expression levels of the anti- and proinflammatory cytokines, lymphocyte recognition receptors and pattern recognition receptors were studied with reverse transcription-polymerase chain reaction (RT-PCR) in 14 children with JIA and 12 controls. All subjects had various gastrointestinal (GI) symptoms indicating endoscopic examinations, but eventually were not diagnosed with GI disease. In JIA patients, the expression of HLA-DR was increased in the crypt epithelial cells and in the epithelial basement membrane of the ileum when compared with the controls. Positive HLA-DR staining in the ileal mucosa was associated with the presence of high clinical disease activity of JIA and low mRNA expression of anti-inflammatory mediators, such as forkhead box protein P3 (FoxP3), glucocorticoid-induced tumour necrosis factor receptor-related protein (GITR) and transforming growth factor (TGF)-beta. Low ileal expression of interleukin (IL)-10, TGF-β, FoxP3, Toll-like receptor 2 (TLR-2) and TLR-4 transcripts correlated significantly with a high clinical disease activity in the JIA patients. The increased HLA-DR expression suggests enhanced intestinal antigen presentation in JIA. A correlation between clinical disease activity and low gene expression of tolerogenic mediators in the ileum supports the hypothesis that a link exists between the gut immune system and JIA.
Collapse
Affiliation(s)
- M Arvonen
- Department of Paediatrics, University of Oulu, Oulu University Hospital, Oulu, Finland
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Kapitein B, Aalberse JA, Klein MR, de Jager W, Hoekstra MO, Knol EF, Prakken BJ. Recognition of self-heat shock protein 60 by T cells from patients with atopic dermatitis. Cell Stress Chaperones 2013; 18:87-95. [PMID: 22869467 PMCID: PMC3508125 DOI: 10.1007/s12192-012-0361-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2012] [Revised: 07/14/2012] [Accepted: 07/16/2012] [Indexed: 12/20/2022] Open
Abstract
Heat shock protein 60 (hsp60) is a highly conserved stress protein and target of self-reactive T cells in various inflammatory diseases. Not much is known about a possible role in atopic disease. As atopic diseases are considered to be the result of a disturbance in the balance between T helper cells type 2 and regulatory T cells, it is of interest to know whether hsp60 acts as a bystander antigen in atopic disease. Our aim was to investigate whether hsp60 is involved in the chronicity of inflammation of atopic dermatitis (AD). We studied the expression of hsp60 in skin tissue of adults with AD by immunohistochemistry. Peripheral blood mononuclear cells (PBMC) of children with AD were cultured with hsp60 and proliferative responses, cytokine secretion, surface markers, and functional assays were compared to responses of PBMC of healthy controls (HC). Hsp60 was detected more in lesional skin of AD patients compared to nonlesional skin. Furthermore, PBMC of children with AD proliferated more strongly in response to hsp60 compared to HC. hsp60-reactive T cells of atopic children produced high levels of IFNγ and low levels of IL-10. In vitro activation with hsp60 leads to the induction of CD4(+)CD25(bright) T cells expressing FOXP3 in both HC as well as in atopic children. However, despite their regulatory phenotype, hsp60-induced CD4(+)CD25(bright)CD127(-)FOXP3(+) T cells of AD patients were incapable of suppressing effector T cells in vitro. hsp60 is recognized by proinflammatory (IFNγ high, IL-10 low) T cells in atopic patients and is more present in lesional AD skin. This suggests that hsp60-specific T cell responses contribute to local inflammation in AD.
Collapse
Affiliation(s)
- Berber Kapitein
- Department of General Paediatrics, Wilhelmina Children’s Hospital, Utrecht, Netherlands
- Department of Paediatric Immunology, University Medical Centre Utrecht, Wilhelmina Children’s Hospital, KC 01.069.0, P.O. Box 85090, 3508 AB Utrecht, Netherlands
- Pediatric Intensive Care Unit, Sophia Children’s Hospital, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Joost A. Aalberse
- Department of General Paediatrics, Wilhelmina Children’s Hospital, Utrecht, Netherlands
- Department of Paediatric Immunology, University Medical Centre Utrecht, Wilhelmina Children’s Hospital, KC 01.069.0, P.O. Box 85090, 3508 AB Utrecht, Netherlands
| | - Mark R. Klein
- Department of Paediatric Immunology, University Medical Centre Utrecht, Wilhelmina Children’s Hospital, KC 01.069.0, P.O. Box 85090, 3508 AB Utrecht, Netherlands
| | - Wilco de Jager
- Department of Paediatric Immunology, University Medical Centre Utrecht, Wilhelmina Children’s Hospital, KC 01.069.0, P.O. Box 85090, 3508 AB Utrecht, Netherlands
| | - Maarten O. Hoekstra
- Department of General Paediatrics, Wilhelmina Children’s Hospital, Utrecht, Netherlands
| | - Edward F. Knol
- Department of Dermatology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Berent J. Prakken
- Department of Paediatric Immunology, University Medical Centre Utrecht, Wilhelmina Children’s Hospital, KC 01.069.0, P.O. Box 85090, 3508 AB Utrecht, Netherlands
| |
Collapse
|
20
|
Billetta R, Ghahramani N, Morrow O, Prakken B, de Jong H, Meschter C, Lanza P, Albani S. Epitope-specific immune tolerization ameliorates experimental autoimmune encephalomyelitis. Clin Immunol 2012; 145:94-101. [DOI: 10.1016/j.clim.2012.08.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2012] [Revised: 07/27/2012] [Accepted: 08/07/2012] [Indexed: 01/04/2023]
|
21
|
van Eden W, Spiering R, Broere F, van der Zee R. A case of mistaken identity: HSPs are no DAMPs but DAMPERs. Cell Stress Chaperones 2012; 17:281-92. [PMID: 22139593 PMCID: PMC3312964 DOI: 10.1007/s12192-011-0311-5] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Accepted: 11/15/2011] [Indexed: 01/19/2023] Open
Abstract
Until recently, the immune system was seen solely as a defense system with its primary task being the elimination of unwanted microbial invaders. Currently, however, the functional significance of the immune system has obtained a much wider perspective, to include among others the maintenance and restoration of homeostasis following tissue damage. In this latter aspect, there is a growing interest in the identification of molecules involved, such as the so-called danger or damage-associated molecular patterns (DAMPs), also called alarmins. Since heat shock proteins are archetypical molecules produced under stressful conditions, such as tissue damage or inflammation, they are frequently mentioned as prime examples of DAMPs (Bianchi, J Leukoc Biol 81:1-5, 2007; Kono and Rock, Nat Rev Immunol 8:279-289, 2008; Martin-Murphy et al., Toxicol Lett 192:387-394, 2010). See for instance also a recent review (Chen and Nunez, Science 298:1395-1401, 2010). Contrary to this description, we recently presented some of the arguments against a role of heat shock protein as DAMPs (Broere et al., Nat Rev Immunol 11:565-c1, 2011). With this perspective and reflection article, we hope to elaborate on this debate and provide additional thoughts to further ignite this discussion on this critical and evolving issue.
Collapse
Affiliation(s)
- Willem van Eden
- Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, Netherlands.
| | | | | | | |
Collapse
|
22
|
Abstract
Progress in understanding the cellular and molecular mechanisms of rheumatoid arthritis (RA), together with the availability of new therapies, has changed the way we think about RA. The paradigm shift in RA therapy has been from controlling symptoms to controlling the disease process with the abrogation of inflammation. Challenges that are still unresolved include the issues in disease prevention, treatment specificity to restore tolerance, approaches to facilitate tissue repair, and treatment optimization to fit the individual patient's disease phenotype and comorbidity context. This review summarizes the pathogenesis-related rationales for the current therapeutic strategies in RA and for emerging therapies and potential approaches to restoring immune tolerance in RA.
Collapse
|
23
|
Shields AM, Panayi GS, Corrigall VM. A New-Age for Biologic Therapies: Long-Term Drug-Free Therapy with BiP? Front Immunol 2012; 3:17. [PMID: 22566902 PMCID: PMC3342250 DOI: 10.3389/fimmu.2012.00017] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2011] [Accepted: 02/01/2012] [Indexed: 12/21/2022] Open
Abstract
Heat shock proteins (HSPs) and other members of the much broader stress protein family have been shown to play important roles in coordinating multiple phases of immunological reactions; from facilitating immunological recognition, to promoting and regulating immunological responses and finally augmenting the resolution of inflammation and return to immunological homeostasis. In this review, we consider the challenges facing the stress protein field as we enter 2012; in particular we consider the role that HSPs and stress proteins may play in the initiation and termination of immunological responses. Special attention is afforded to the resolution-associated molecular pattern, binding immunoglobulin protein (BiP, also known as glucose regulated protein-78). We review the evidence that resolution-promoting proteins such as BiP may herald a new generation of biologics for inflammatory disease and reflect on the challenges of achieving clinical remission in rheumatoid arthritis with novel therapeutics and correlating clinical remission with immunological parameters of resolution of inflammation.
Collapse
Affiliation(s)
- Adrian M Shields
- Department of Academic Rheumatology, Centre for Molecular and Cellular Biology of Inflammation, King's College London London, UK
| | | | | |
Collapse
|
24
|
Takakubo Y, Konttinen YT. Immune-regulatory mechanisms in systemic autoimmune and rheumatic diseases. Clin Dev Immunol 2011; 2012:941346. [PMID: 22110541 PMCID: PMC3207139 DOI: 10.1155/2012/941346] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2011] [Revised: 08/06/2011] [Accepted: 08/18/2011] [Indexed: 02/07/2023]
Abstract
Systemic autoimmune and rheumatic diseases (SAIRDs) are thought to develop due to the failure of autoimmune regulation and tolerance. Current therapies, such as biologics, have improved the clinical results of SAIRDs; however, they are not curative treatments. Recently, new discoveries have been made in immune tolerance and inflammation, such as tolerogenic dendritic cells, regulatory T and B cells, Th 17 cells, inflammatory and tolerogenic cytokines, and intracellular signaling pathways. They lay the foundation for the next generation of the therapies beyond the currently used biologic therapies. New drugs should target the core processes involved in disease mechanisms with the aim to attain complete cure combined with safety and low costs compared to the biologic agents. Re-establishment of autoimmune regulation and tolerance in SAIRDs by the end of the current decade should be the final and realistic target.
Collapse
Affiliation(s)
- Yuya Takakubo
- Department of Medicine, Biomedicum Helsinki, University of Helsinki, PO Box 700, Haartmaninkatu 8, 00029 HUS, Finland.
| | | |
Collapse
|
25
|
Pham CTN. Nanotherapeutic approaches for the treatment of rheumatoid arthritis. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2011; 3:607-19. [PMID: 21837725 DOI: 10.1002/wnan.157] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Rheumatoid arthritis (RA) is a common inflammatory disease characterized by progressive bone and cartilage destruction, resulting in severe functional limitations, shortened lifespan, and increased mortality rates. Recent advances and new treatment approaches have significantly delayed disease progression and improved the quality of life for many patients. Yet few patients attain or can be maintained in disease remission without continuous immunosuppressive therapy. In addition, a sizable portion of patients also fails to respond or eventually develops tolerance to current therapies. Thus there is a continued need for the development of new therapeutic strategies for the treatment of RA. Unlike conventional drugs, nanosystems are designed to deliver therapeutic agents specifically to the site of inflammation, therefore avoiding potential systemic and off-target unwanted effects. They allow investigators to consider or reconsider therapeutic agents that were previously deemed too toxic to deliver through a systemic route. This article reviews recent nanotechnology-based strategies that are being developed for the treatment of inflammatory arthritis.
Collapse
Affiliation(s)
- Christine T N Pham
- Division of Rheumatology, Washington University School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|