1
|
Quero FB, Troncoso-Bravo T, Farías MA, Kalergis AM. Cell-Based Therapeutic Strategies for Autoimmune Diseases. Immunotargets Ther 2025; 14:501-514. [PMID: 40322732 PMCID: PMC12047289 DOI: 10.2147/itt.s513629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 04/15/2025] [Indexed: 05/08/2025] Open
Abstract
Currently, the management of autoimmune disorders still being a challenge in terms of safety, efficiency, and specificity. Cell-based therapeutic strategies have emerged as a novel approach for autoimmune disease treatment, employing different cell therapy platforms, including tolerogenic dendritic cells, regulatory T cells, conventional and regulatory chimeric antigen receptor-T cells, mesenchymal and hematopoietic stem cells, each with their biological features. Here, we discuss the different cell therapy platforms, their immunological mechanisms of action, their therapeutic potential and benefits in autoimmune diseases, and challenges related to their production, scaling up, risks, and patient safety.
Collapse
Affiliation(s)
- Francisco B Quero
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Tays Troncoso-Bravo
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Mónica A Farías
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
2
|
Yu XH, Lu HM, Li J, Su MZ, Li XM, Jin Y. Association between 25(OH) vitamin D and multiple sclerosis: cohort, shared genetics, and Causality. Nutr J 2024; 23:151. [PMID: 39616386 PMCID: PMC11608472 DOI: 10.1186/s12937-024-01059-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 11/26/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND Multiple Sclerosis (MS), an autoimmune disorder causing demyelination and neurological damage, has been linked to 25-hydroxyvitamin D (25OHD) levels, suggesting its role in immune response and MS onset. This study used GWAS datasets to investigate genetic associations between 25OHD and MS. METHODS We utilized a large-scale prospective cohort to evaluate serum 25OHD levels and MS risk. Linkage Disequilibrium Score Regression (LDSC) assessed genetic correlations between 25OHD levels and MS. Cross-trait genome-wide pleiotropy analysis revealed shared genetic loci. MAGMA analysis identified pleiotropic genes, enriched tissues, and gene sets. Stratified LDSC estimated tissue-specific and cell-specific heritability enrichment, and multi-trait co-localization analysis identified shared immune cell subsets. Bidirectional Mendelian Randomization (MR) assessed the causal association between 25OHD and MS risk. RESULTS The observational study found a nonlinear relationship between 25OHD levels and MS risk, with the lowest quartile showing significant risk elevation. Our findings revealed shared genetic structure between 25OHD levels and MS, suggesting a common biological pathway involving immune function and CNS integrity. We found 24 independent loci shared between 25OHD levels and MS risk, enriched in brain tissues and involved in pathways like LDL, HDL, and TG metabolism. Four loci (6p24.3, 6p22.2, 12q14.1, and 19p13.2) had strong co-localization evidence, with mapped genes as potential drug targets. Bidirectional MR analysis supported a causal effect of 25OHD levels on MS risk, suggesting 25OHD supplementation could modulate MS risk. CONCLUSION This study reveals the complex relationship between 25OHD levels and MS, indicating that higher levels are not always advantageous and recommending moderation in supplementation. We identified SMARCA4 as a potential therapeutic target and detailed key pathways influencing this interaction.
Collapse
Affiliation(s)
- Xing-Hao Yu
- The Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, 213000, China
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215123, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, 215123, China
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Hui-Min Lu
- Department of Outpatient and Emergency, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215123, China
| | - Jun Li
- The Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, 213000, China
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China
- Department of Pharmacy, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu, 213000, China
| | - Ming-Zhu Su
- The Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, 213000, China
- Department of Good Clinical Practice, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu, 213000, China
| | - Xiao-Min Li
- The Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, 213000, China.
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China.
- Department of Pharmacy, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu, 213000, China.
| | - Yi Jin
- The Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, 213000, China.
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou, Jiangsu, 221002, China.
- Department of Pharmacy, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu, 213000, China.
| |
Collapse
|
3
|
Fondelli F, Willemyns J, Domenech-Garcia R, Mansilla MJ, Godoy-Tena G, Ferreté-Bonastre AG, Agúndez-Moreno A, Presas-Rodriguez S, Ramo-Tello C, Ballestar E, Martínez-Cáceres E. Targeting aryl hydrocarbon receptor functionally restores tolerogenic dendritic cells derived from patients with multiple sclerosis. J Clin Invest 2024; 134:e178949. [PMID: 39287981 PMCID: PMC11527446 DOI: 10.1172/jci178949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 09/11/2024] [Indexed: 09/19/2024] Open
Abstract
Multiple sclerosis (MS) is a chronic disease characterized by dysregulated self-reactive immune responses that damage the neurons' myelin sheath, leading to progressive disability. The primary therapeutic option, immunosuppressants, inhibits pathogenic anti-myelin responses but depresses the immune system. Antigen-specific monocyte-derived autologous tolerogenic dendritic cells (tolDCs) offer alternative therapeutic approaches to restore tolerance to autoantigens without causing generalized immunosuppression. However, immune dysregulation in MS could impact the properties of the monocytes used as starting material for this cell therapy. Here, we characterized CD14+ monocytes, mature dendritic cells, and vitamin D3-tolDCs (VitD3-tolDCs) from active, treatment-naive MS patients and healthy donors (HDs). Using multiomics, we identified a switch in these cell types toward proinflammatory features characterized by alterations in the aryl hydrocarbon receptor (AhR) and NF-κB pathways. MS patient-derived VitD3-tolDCs showed reduced tolerogenic properties compared with those from HDs, which were fully restored through direct AhR agonism and by use of in vivo or in vitro dimethyl fumarate (DMF) supplementation. Additionally, in the experimental autoimmune encephalomyelitis mouse model, combined therapy of DMF and VitD3-tolDCs was more efficient than monotherapies in reducing the clinical score of mice. We propose that a combined therapy with DMF and VitD3-tolDCs offers enhanced therapeutic potential in treating MS.
Collapse
MESH Headings
- Humans
- Dendritic Cells/immunology
- Receptors, Aryl Hydrocarbon/immunology
- Receptors, Aryl Hydrocarbon/agonists
- Receptors, Aryl Hydrocarbon/metabolism
- Animals
- Mice
- Female
- Male
- Immune Tolerance
- Multiple Sclerosis/immunology
- Multiple Sclerosis/pathology
- Multiple Sclerosis/therapy
- Multiple Sclerosis/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/therapy
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Adult
- Middle Aged
- Monocytes/immunology
- Monocytes/metabolism
- NF-kappa B/metabolism
- NF-kappa B/immunology
- Cholecalciferol/pharmacology
- Basic Helix-Loop-Helix Transcription Factors/immunology
- Basic Helix-Loop-Helix Transcription Factors/metabolism
- Basic Helix-Loop-Helix Transcription Factors/genetics
Collapse
Affiliation(s)
- Federico Fondelli
- Immunology Division, Laboratori Clínic de la Metropolitana Nord, Germans Trias i Pujol University Hospital and Research Institute, Campus Can Ruti, Badalona, Spain
- Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Jana Willemyns
- Immunology Division, Laboratori Clínic de la Metropolitana Nord, Germans Trias i Pujol University Hospital and Research Institute, Campus Can Ruti, Badalona, Spain
- Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Roger Domenech-Garcia
- Immunology Division, Laboratori Clínic de la Metropolitana Nord, Germans Trias i Pujol University Hospital and Research Institute, Campus Can Ruti, Badalona, Spain
- Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Maria José Mansilla
- Immunology Division, Laboratori Clínic de la Metropolitana Nord, Germans Trias i Pujol University Hospital and Research Institute, Campus Can Ruti, Badalona, Spain
| | - Gerard Godoy-Tena
- Epigenetics and Immune Disease Group, Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Spain
| | - Anna G. Ferreté-Bonastre
- Epigenetics and Immune Disease Group, Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Spain
| | - Alex Agúndez-Moreno
- Immunology Division, Laboratori Clínic de la Metropolitana Nord, Germans Trias i Pujol University Hospital and Research Institute, Campus Can Ruti, Badalona, Spain
- Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Silvia Presas-Rodriguez
- Multiple Sclerosis Unit, Department of Neurosciences, Germans Trias i Pujol University Hospital, Badalona, Spain
- Department of Medicine, Campus Bellaterra, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Cristina Ramo-Tello
- Multiple Sclerosis Unit, Department of Neurosciences, Germans Trias i Pujol University Hospital, Badalona, Spain
- Department of Medicine, Campus Bellaterra, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Esteban Ballestar
- Epigenetics and Immune Disease Group, Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Spain
- Epigenetics in Inflammatory and Metabolic Diseases Laboratory, Health Science Center (HSC), East China Normal University (ECNU), Shanghai, China
| | - Eva Martínez-Cáceres
- Immunology Division, Laboratori Clínic de la Metropolitana Nord, Germans Trias i Pujol University Hospital and Research Institute, Campus Can Ruti, Badalona, Spain
- Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| |
Collapse
|
4
|
Song Y, Li J, Wu Y. Evolving understanding of autoimmune mechanisms and new therapeutic strategies of autoimmune disorders. Signal Transduct Target Ther 2024; 9:263. [PMID: 39362875 PMCID: PMC11452214 DOI: 10.1038/s41392-024-01952-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 07/09/2024] [Accepted: 08/07/2024] [Indexed: 10/05/2024] Open
Abstract
Autoimmune disorders are characterized by aberrant T cell and B cell reactivity to the body's own components, resulting in tissue destruction and organ dysfunction. Autoimmune diseases affect a wide range of people in many parts of the world and have become one of the major concerns in public health. In recent years, there have been substantial progress in our understanding of the epidemiology, risk factors, pathogenesis and mechanisms of autoimmune diseases. Current approved therapeutic interventions for autoimmune diseases are mainly non-specific immunomodulators and may cause broad immunosuppression that leads to serious adverse effects. To overcome the limitations of immunosuppressive drugs in treating autoimmune diseases, precise and target-specific strategies are urgently needed. To date, significant advances have been made in our understanding of the mechanisms of immune tolerance, offering a new avenue for developing antigen-specific immunotherapies for autoimmune diseases. These antigen-specific approaches have shown great potential in various preclinical animal models and recently been evaluated in clinical trials. This review describes the common epidemiology, clinical manifestation and mechanisms of autoimmune diseases, with a focus on typical autoimmune diseases including multiple sclerosis, type 1 diabetes, rheumatoid arthritis, systemic lupus erythematosus, and sjögren's syndrome. We discuss the current therapeutics developed in this field, highlight the recent advances in the use of nanomaterials and mRNA vaccine techniques to induce antigen-specific immune tolerance.
Collapse
Affiliation(s)
- Yi Song
- Institute of Immunology, PLA, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jian Li
- Chongqing International Institute for Immunology, Chongqing, China.
| | - Yuzhang Wu
- Institute of Immunology, PLA, Third Military Medical University (Army Medical University), Chongqing, China.
- Chongqing International Institute for Immunology, Chongqing, China.
| |
Collapse
|
5
|
Li V, Binder MD, Purcell AW, Kilpatrick TJ. Antigen-specific immunotherapy via delivery of tolerogenic dendritic cells for multiple sclerosis. J Neuroimmunol 2024; 390:578347. [PMID: 38663308 DOI: 10.1016/j.jneuroim.2024.578347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 03/22/2024] [Accepted: 04/17/2024] [Indexed: 05/13/2024]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease of the central nervous system resulting from loss of immune tolerance. Many disease-modifying therapies for MS have broad immunosuppressive effects on peripheral immune cells, but this can increase risks of infection and attenuate vaccine-elicited immunity. A more targeted approach is to re-establish immune tolerance in an autoantigen-specific manner. This review discusses methods to achieve this, focusing on tolerogenic dendritic cells. Clinical trials in other autoimmune diseases also provide learnings with regards to clinical translation of this approach, including identification of autoantigen(s), selection of appropriate patients and administration route and frequency.
Collapse
Affiliation(s)
- Vivien Li
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3010, Australia; Department of Neurology, The Royal Melbourne Hospital, Melbourne, Australia.
| | - Michele D Binder
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3010, Australia
| | - Anthony W Purcell
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Australia
| | - Trevor J Kilpatrick
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville 3010, Australia; Department of Neurology, The Royal Melbourne Hospital, Melbourne, Australia
| |
Collapse
|
6
|
Dao Nyesiga G, Pool L, Englezou PC, Hylander T, Ohlsson L, Appelgren D, Sundstedt A, Tillerkvist K, Romedahl HR, Wigren M. Tolerogenic dendritic cells generated in vitro using a novel protocol mimicking mucosal tolerance mechanisms represent a potential therapeutic cell platform for induction of immune tolerance. Front Immunol 2023; 14:1045183. [PMID: 37901231 PMCID: PMC10613069 DOI: 10.3389/fimmu.2023.1045183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 08/25/2023] [Indexed: 10/31/2023] Open
Abstract
Dendritic cells (DCs) are mediators between innate and adaptive immunity and vital in initiating and modulating antigen-specific immune responses. The most important site for induction of tolerance is the gut mucosa, where TGF-β, retinoic acid, and aryl hydrocarbon receptors collaborate in DCs to induce a tolerogenic phenotype. To mimic this, a novel combination of compounds - the synthetic aryl hydrocarbon receptor (AhR) agonist IGN-512 together with TGF-β and retinoic acid - was developed to create a platform technology for induction of tolerogenic DCs intended for treatment of several conditions caused by unwanted immune activation. These in vitro-generated cells, designated ItolDCs, are phenotypically characterized by their low expression of co-stimulatory and activating molecules along with high expression of tolerance-associated markers such as ILT3, CD103, and LAP, and a weak pro-inflammatory cytokine profile. When co-cultured with T cells and/or B cells, ItolDC-cultures contain higher frequencies of CD25+Foxp3+ regulatory T cells (Tregs), CD49b+LAG3+ 'type 1 regulatory (Tr1) T cells, and IL-10-producing B cells and are less T cell stimulatory compared to cultures with matured DCs. Factor VIII (FVIII) and tetanus toxoid (TT) were used as model antigens to study ItolDC antigen-loading. ItolDCs can take up FVIII, process, and present FVIII peptides on HLA-DR. By loading both ItolDCs and mDCs with TT, antigen-specific T cell proliferation was observed. Cryo-preserved ItolDCs showed a stable tolerogenic phenotype that was maintained after stimulation with LPS, CD40L, or a pro-inflammatory cocktail. Moreover, exposure to other immune cells did not negatively impact ItolDCs' expression of tolerogenic markers. In summary, a novel protocol was developed supporting the generation of a stable population of human DCs in vitro that exhibited a tolerogenic phenotype with an ability to increase proportions of induced regulatory T and B cells in mixed cultures. This protocol has the potential to constitute the base of a tolDC platform for inducing antigen-specific tolerance in disorders caused by undesired antigen-specific immune cell activation.
Collapse
Affiliation(s)
- Gillian Dao Nyesiga
- Idogen AB, Lund, Sweden
- Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, Malmö, Sweden
| | | | | | | | - Lars Ohlsson
- Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, Malmö, Sweden
| | - Daniel Appelgren
- Department of Health, Medicine and Caring Sciences, Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| | | | | | | | | |
Collapse
|
7
|
Passeri L, Andolfi G, Bassi V, Russo F, Giacomini G, Laudisa C, Marrocco I, Cesana L, Di Stefano M, Fanti L, Sgaramella P, Vitale S, Ziparo C, Auricchio R, Barera G, Di Nardo G, Troncone R, Gianfrani C, Annoni A, Passerini L, Gregori S. Tolerogenic IL-10-engineered dendritic cell-based therapy to restore antigen-specific tolerance in T cell mediated diseases. J Autoimmun 2023; 138:103051. [PMID: 37224733 DOI: 10.1016/j.jaut.2023.103051] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 02/06/2023] [Accepted: 04/21/2023] [Indexed: 05/26/2023]
Abstract
Tolerogenic dendritic cells play a critical role in promoting antigen-specific tolerance via dampening of T cell responses, induction of pathogenic T cell exhaustion and antigen-specific regulatory T cells. Here we efficiently generate tolerogenic dendritic cells by genetic engineering of monocytes with lentiviral vectors co-encoding for immunodominant antigen-derived peptides and IL-10. These transduced dendritic cells (designated DCIL-10/Ag) secrete IL-10 and efficiently downregulate antigen-specific CD4+ and CD8+ T cell responses from healthy subjects and celiac disease patients in vitro. In addition, DCIL-10/Ag induce antigen-specific CD49b+LAG-3+ T cells, which display the T regulatory type 1 (Tr1) cell gene signature. Administration of DCIL-10/Ag resulted in the induction of antigen-specific Tr1 cells in chimeric transplanted mice and the prevention of type 1 diabetes in pre-clinical disease models. Subsequent transfer of these antigen-specific T cells completely prevented type 1 diabetes development. Collectively these data indicate that DCIL-10/Ag represent a platform to induce stable antigen-specific tolerance to control T-cell mediated diseases.
Collapse
Affiliation(s)
- Laura Passeri
- Mechanisms of Peripheral Tolerance Unit, San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Grazia Andolfi
- Mechanisms of Peripheral Tolerance Unit, San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Virginia Bassi
- Mechanisms of Peripheral Tolerance Unit, San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy; University of Rome Tor Vergata, Via Cracovia 50, 00133, Rome, Italy
| | - Fabio Russo
- Mechanisms of Peripheral Tolerance Unit, San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Giorgia Giacomini
- Mechanisms of Peripheral Tolerance Unit, San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Cecilia Laudisa
- Mechanisms of Peripheral Tolerance Unit, San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Ilaria Marrocco
- Mechanisms of Peripheral Tolerance Unit, San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Luca Cesana
- Mechanisms of Peripheral Tolerance Unit, San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Marina Di Stefano
- Department of Paediatrics, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Lorella Fanti
- Gastroenterology and Gastrointestinal Endoscopy Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Paola Sgaramella
- Department of Paediatrics, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Serena Vitale
- Institute of Biochemistry and Cell Biology, CNR, via P.Castellino 11, 80131, Naples, Italy
| | - Chiara Ziparo
- NESMOS Department, School of Medicine and Psychology, Sapienza University of Rome, Sant'Andrea University Hospital, Via di Grottarossa 1035, 00189, Rome, Italy
| | - Renata Auricchio
- European Laboratory for the Investigation of Food Induced Diseases (ELFID), Department of Translational Medical Science, Section of Pediatrics, Via Pansini 5, 80131, University Federico II, Naples, Italy
| | - Graziano Barera
- Department of Paediatrics, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Giovanni Di Nardo
- NESMOS Department, School of Medicine and Psychology, Sapienza University of Rome, Sant'Andrea University Hospital, Via di Grottarossa 1035, 00189, Rome, Italy
| | - Riccardo Troncone
- European Laboratory for the Investigation of Food Induced Diseases (ELFID), Department of Translational Medical Science, Section of Pediatrics, Via Pansini 5, 80131, University Federico II, Naples, Italy
| | - Carmen Gianfrani
- Institute of Biochemistry and Cell Biology, CNR, via P.Castellino 11, 80131, Naples, Italy
| | - Andrea Annoni
- Mechanisms of Peripheral Tolerance Unit, San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Laura Passerini
- Mechanisms of Peripheral Tolerance Unit, San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Silvia Gregori
- Mechanisms of Peripheral Tolerance Unit, San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy.
| |
Collapse
|
8
|
Zhang X, Liu J, Cheng Y, Chen K, Chen Y, Zhu H, Li Z, Liu S, Cao X. Metabolic enzyme Suclg2 maintains tolerogenicity of regulatory dendritic cells diffDCs by suppressing Lactb succinylation. J Autoimmun 2023; 138:103048. [PMID: 37216870 DOI: 10.1016/j.jaut.2023.103048] [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: 12/05/2022] [Revised: 04/11/2023] [Accepted: 04/18/2023] [Indexed: 05/24/2023]
Abstract
Metabolic reprogramming plays a pivotal role in the differentiation and function of immune cells including dendritic cells (DCs). Regulatory DCs can be generated in regional tissue niches like splenic stroma and act as an important part of stromal control of immune response for the maintenance of immune tolerance. However, the metabolic alterations during splenic stroma-driven regulatory DCs differentiation and the metabolic enzyme involved in regulatory DCs function remain poorly understood. By combining metabolomic, transcriptomic, and functional investigations of mature DCs (maDCs) and diffDCs (regulatory DCs differentiated from activated mature DCs through coculturing with splenic stroma), here we identified succinate-CoA ligase subunit beta Suclg2 as a key metabolic enzyme that reprograms the proinflammatory status of mature DCs into a tolerogenic phenotype via preventing NF-κB signaling activation. diffDCs downregulate succinic acid levels and increase the Suclg2 expression along with their differentiation from mature DCs. Suclg2-interference impaired the tolerogenic function of diffDCs in inducing T cell apoptosis and enhanced activation of NF-κB signaling and expression of inflammatory genes CD40, Ccl5, and Il12b in diffDCs. Furthermore, we identified Lactb as a new positive regulator of NF-κB signaling in diffDCs whose succinylation at the lysine 288 residue was inhibited by Suclg2. Our study reveals that the metabolic enzyme Suclg2 is required to maintain the immunoregulatory function of diffDCs, adding mechanistic insights into the metabolic regulation of DC-based immunity and tolerance.
Collapse
Affiliation(s)
- Xiaomin Zhang
- National Key Laboratory of Medical Immunology, Institute of Immunology, Second Military Medical University, Shanghai, 200433, China
| | - Juan Liu
- National Key Laboratory of Medical Immunology, Institute of Immunology, Second Military Medical University, Shanghai, 200433, China.
| | - Yujie Cheng
- National Key Laboratory of Medical Immunology, Institute of Immunology, Second Military Medical University, Shanghai, 200433, China
| | - Kun Chen
- Department of Immunology, Center for Immunotherapy, Institute of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Yali Chen
- Department of Immunology, Center for Immunotherapy, Institute of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Ha Zhu
- National Key Laboratory of Medical Immunology, Institute of Immunology, Second Military Medical University, Shanghai, 200433, China
| | - Zhiqing Li
- National Key Laboratory of Medical Immunology, Institute of Immunology, Second Military Medical University, Shanghai, 200433, China
| | - Shuxun Liu
- National Key Laboratory of Medical Immunology, Institute of Immunology, Second Military Medical University, Shanghai, 200433, China
| | - Xuetao Cao
- National Key Laboratory of Medical Immunology, Institute of Immunology, Second Military Medical University, Shanghai, 200433, China; Department of Immunology, Center for Immunotherapy, Institute of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100005, China; Frontier Research Center for Cell Response, Institute of Immunology, College of Life Sciences, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
9
|
Immunity orchestrates a bridge in gut-brain axis of neurodegenerative diseases. Ageing Res Rev 2023; 85:101857. [PMID: 36669690 DOI: 10.1016/j.arr.2023.101857] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 01/15/2023] [Accepted: 01/15/2023] [Indexed: 01/18/2023]
Abstract
Neurodegenerative diseases, in particular for Alzheimer's disease (AD), Parkinson's disease (PD) and Multiple sclerosis (MS), are a category of diseases with progressive loss of neuronal structure or function (encompassing neuronal death) leading to neuronal dysfunction, whereas the underlying pathogenesis remains to be clarified. As the microbiological ecosystem of the intestinal microbiome serves as the second genome of the human body, it is strongly implicated as an essential element in the initiation and/or progression of neurodegenerative diseases. Nevertheless, the precise underlying principles of how the intestinal microflora impact on neurodegenerative diseases via gut-brain axis by modulating the immune function are still poorly characterized. Consequently, an overview of initiating the development of neurodegenerative diseases and the contribution of intestinal microflora on immune function is discussed in this review.
Collapse
|
10
|
Xue E, Minniti A, Alexander T, Del Papa N, Greco R, on behalf of The Autoimmune Diseases Working Party (ADWP) of the European Society for Blood and Marrow Transplantation (EBMT). Cellular-Based Therapies in Systemic Sclerosis: From Hematopoietic Stem Cell Transplant to Innovative Approaches. Cells 2022; 11:3346. [PMID: 36359742 PMCID: PMC9658618 DOI: 10.3390/cells11213346] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 10/19/2022] [Accepted: 10/19/2022] [Indexed: 08/28/2023] Open
Abstract
Systemic sclerosis (SSc) is a systemic disease characterized by autoimmune responses, vasculopathy and tissue fibrosis. The pathogenic mechanisms involve a wide range of cells and soluble factors. The complexity of interactions leads to heterogeneous clinical features in terms of the extent, severity, and rate of progression of skin fibrosis and internal organ involvement. Available disease-modifying drugs have only modest effects on halting disease progression and may be associated with significant side effects. Therefore, cellular therapies have been developed aiming at the restoration of immunologic self-tolerance in order to provide durable remissions or to foster tissue regeneration. Currently, SSc is recommended as the 'standard indication' for autologous hematopoietic stem cell transplantation by the European Society for Blood and Marrow Transplantation. This review provides an overview on cellular therapies in SSc, from pre-clinical models to clinical applications, opening towards more advanced cellular therapies, such as mesenchymal stem cells, regulatory T cells and potentially CAR-T-cell therapies.
Collapse
Affiliation(s)
- Elisabetta Xue
- Hematopoietic and Bone Marrow Transplant Unit, San Raffaele Hospital, 20132 Milan, Italy
| | - Antonina Minniti
- Department of Rheumatology, ASST G. Pini-CTO, 20122 Milan, Italy
| | - Tobias Alexander
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Rheumatology and Clinical Immunology, 10117 Berlin, Germany
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, 10117 Berlin, Germany
| | | | - Raffaella Greco
- Hematopoietic and Bone Marrow Transplant Unit, San Raffaele Hospital, 20132 Milan, Italy
| | | |
Collapse
|
11
|
Galoppin M, Kari S, Soldati S, Pal A, Rival M, Engelhardt B, Astier A, Thouvenot E. Full spectrum of vitamin D immunomodulation in multiple sclerosis: mechanisms and therapeutic implications. Brain Commun 2022; 4:fcac171. [PMID: 35813882 PMCID: PMC9260308 DOI: 10.1093/braincomms/fcac171] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 05/03/2022] [Accepted: 06/28/2022] [Indexed: 11/17/2022] Open
Abstract
Vitamin D deficiency has been associated with the risk of multiple sclerosis, disease activity and progression. Results from in vitro experiments, animal models and analysis of human samples from randomized controlled trials provide comprehensive data illustrating the pleiotropic actions of Vitamin D on the immune system. They globally result in immunomodulation by decreasing differentiation of effector T and B cells while promoting regulatory subsets. Vitamin D also modulates innate immune cells such as macrophages, monocytes and dendritic cells, and acts at the level of the blood–brain barrier reducing immune cell trafficking. Vitamin D exerts additional activity within the central nervous system reducing microglial and astrocytic activation. The immunomodulatory role of Vitamin D detected in animal models of multiple sclerosis has suggested its potential therapeutic use for treating multiple sclerosis. In this review, we focus on recent published data describing the biological effects of Vitamin D in animal models of multiple sclerosis on immune cells, blood–brain barrier function, activation of glial cells and its potential neuroprotective effects. Based on the current knowledge, we also discuss optimization of therapeutic interventions with Vitamin D in patients with multiple sclerosis, as well as new technologies allowing in-depth analysis of immune cell regulations by vitamin D.
Collapse
Affiliation(s)
- Manon Galoppin
- IGF, University Montpellier, CNRS, INSERM , Montpellier , France
| | - Saniya Kari
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291 – CNRS UMR5051 – Université Toulouse III , 31024 Toulouse cedex 3 , France
| | - Sasha Soldati
- Theodor Kocher Institute, University of Bern , Bern , Switzerland
| | - Arindam Pal
- Theodor Kocher Institute, University of Bern , Bern , Switzerland
| | - Manon Rival
- IGF, University Montpellier, CNRS, INSERM , Montpellier , France
- Department of Neurology, Nîmes University Hospital, University Montpellier , Nîmes , France
| | | | - Anne Astier
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291 – CNRS UMR5051 – Université Toulouse III , 31024 Toulouse cedex 3 , France
| | - Eric Thouvenot
- IGF, University Montpellier, CNRS, INSERM , Montpellier , France
- Department of Neurology, Nîmes University Hospital, University Montpellier , Nîmes , France
| |
Collapse
|
12
|
Metabolic Reprogramming of Innate Immune Cells as a Possible Source of New Therapeutic Approaches in Autoimmunity. Cells 2022; 11:cells11101663. [PMID: 35626700 PMCID: PMC9140143 DOI: 10.3390/cells11101663] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 05/05/2022] [Accepted: 05/13/2022] [Indexed: 11/19/2022] Open
Abstract
Immune cells undergo different metabolic pathways or immunometabolisms to interact with various antigens. Immunometabolism links immunological and metabolic processes and is critical for innate and adaptive immunity. Although metabolic reprogramming is necessary for cell differentiation and proliferation, it may mediate the imbalance of immune homeostasis, leading to the pathogenesis and development of some diseases, such as autoimmune diseases. Here, we discuss the effects of metabolic changes in autoimmune diseases, exerted by the leading actors of innate immunity, and their role in autoimmunity pathogenesis, suggesting many immunotherapeutic approaches.
Collapse
|
13
|
Zorzella-Pezavento SFG, Mimura LAN, Denadai MB, de Souza WDF, Fraga-Silva TFDC, Sartori A. Is there a window of opportunity for the therapeutic use of vitamin D in multiple sclerosis? Neural Regen Res 2022; 17:1945-1954. [PMID: 35142671 PMCID: PMC8848597 DOI: 10.4103/1673-5374.335139] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Multiple sclerosis is an autoimmune treatable but not curable disease. There are a multiplicity of medications for multiple sclerosis therapy, including a class entitled disease-modifying drugs that are mainly indicated to reduce the number and severity of disease relapses. Not all patients respond well to these therapies, and minor to severe adverse effects have been reported. Vitamin D, called sunshine vitamin, is being studied as a possible light at the end of the tunnel. In this review, we recapitulated the similar immunopathogenesis of multiple sclerosis and experimental autoimmune encephalomyelitis, the immunomodulatory and neuroprotective potential of vitamin D and the state-of-art concerning its supplementation to multiple sclerosis patients. Finally, based on our and other groups’ experimental findings, we analyzed the need to consider the relevance of the route and the different time-point administration aspects for a more rational indication of this vitamin to multiple sclerosis patients.
Collapse
Affiliation(s)
| | - Luiza Ayumi Nishiyama Mimura
- Department of Chemical and Biological Sciences, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Marina Bonifácio Denadai
- Department of Chemical and Biological Sciences, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - William Danilo Fernandes de Souza
- Department of Chemical and Biological Sciences, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | | | - Alexandrina Sartori
- Department of Chemical and Biological Sciences, Institute of Biosciences, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| |
Collapse
|
14
|
Mi Y, Han J, Zhu J, Jin T. Role of the PD-1/PD-L1 Signaling in Multiple Sclerosis and Experimental Autoimmune Encephalomyelitis: Recent Insights and Future Directions. Mol Neurobiol 2021; 58:6249-6271. [PMID: 34480337 PMCID: PMC8639577 DOI: 10.1007/s12035-021-02495-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 07/12/2021] [Indexed: 12/19/2022]
Abstract
Multiple sclerosis (MS) is an autoimmunity-related chronic demyelination disease of the central nervous system (CNS), causing young disability. Currently, highly specific immunotherapies for MS are still lacking. Programmed cell death 1 (PD-1) is an immunosuppressive co-stimulatory molecule, which is expressed on activated T lymphocytes, B lymphocytes, natural killer cells, and other immune cells. PD-L1, the ligand of PD-1, is expressed on T lymphocytes, B lymphocytes, dendritic cells, and macrophages. PD-1/PD-L1 delivers negative regulatory signals to immune cells, maintaining immune tolerance and inhibiting autoimmunity. This review comprehensively summarizes current insights into the role of PD-1/PD-L1 signaling in MS and its animal model experimental autoimmune encephalomyelitis (EAE). The potentiality of PD-1/PD-L1 as biomarkers or therapeutic targets for MS will also be discussed.
Collapse
Affiliation(s)
- Yan Mi
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021 China
| | - Jinming Han
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021 China
- Present Address: Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jie Zhu
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021 China
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Karolinska University Hospital, Solna, Stockholm, Sweden
| | - Tao Jin
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021 China
| |
Collapse
|
15
|
Quirant-Sánchez B, Mansilla MJ, Navarro-Barriuso J, Presas-Rodríguez S, Teniente-Serra A, Fondelli F, Ramo-Tello C, Martínez-Cáceres E. Combined Therapy of Vitamin D3-Tolerogenic Dendritic Cells and Interferon-β in a Preclinical Model of Multiple Sclerosis. Biomedicines 2021; 9:biomedicines9121758. [PMID: 34944573 PMCID: PMC8698295 DOI: 10.3390/biomedicines9121758] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/18/2021] [Accepted: 11/20/2021] [Indexed: 12/17/2022] Open
Abstract
Autologous antigen-specific therapies based on tolerogenic dendritic cells (tolDC) offer the possibility to treat autoimmune diseases by restoring homeostasis and targeting specifically autoreactive responses. Here, we explore the hypothesis that systemic inflammation occurring in autoimmune diseases, such as multiple sclerosis (MS), can generate a disease-specific environment able to alter the functionality of tolDC. In this context in fact, a combined therapy of tolDC with an immunomodulatory treatment could potentiate the beneficial effect of this antigen-specific cell therapy. For this purpose, we analyzed the efficacy of a combined therapy based on the use of vitamin D3 (VitD3)-tolDC plus interferon beta (IFN-beta) in MS. VitD3-tolDC were generated from healthy donors and MS patients and co-cultured with allogeneic peripheral blood mononuclear cells, in the presence or absence of IFN-beta. In vitro, VitD3-tolDC treatment reduced the percentage of activated T cells and allogeneic proliferation, whereas VitD3-tolDC+IFN-beta treatment enhanced the suppressive ability of VitD3-tolDC and, additionally, induced a shift towards a Th2 profile. To determine the clinical benefit of the combined therapy, C57BL/6-experimental autoimmune encephalomyelitis (EAE)-induced mice were treated with antigen-specific VitD3-tolDC and/or IFN-beta. Treatment of EAE mice with combined therapy ameliorated the disease course compared to each monotherapy. These results suggest that a combined therapy based on antigen-specific VitD3-tolDC and IFN-beta may represent a promising strategy for MS patients.
Collapse
Affiliation(s)
- Bibiana Quirant-Sánchez
- Immunology Division, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Campus Can Ruti, 08916 Badalona, Spain; (B.Q.-S.); (M.J.M.); (J.N.-B.); (A.T.-S.); (F.F.)
- Department of Cellular Biology, Physiology and Immunology, Campus Bellaterra, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
| | - María José Mansilla
- Immunology Division, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Campus Can Ruti, 08916 Badalona, Spain; (B.Q.-S.); (M.J.M.); (J.N.-B.); (A.T.-S.); (F.F.)
- Department of Cellular Biology, Physiology and Immunology, Campus Bellaterra, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
| | - Juan Navarro-Barriuso
- Immunology Division, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Campus Can Ruti, 08916 Badalona, Spain; (B.Q.-S.); (M.J.M.); (J.N.-B.); (A.T.-S.); (F.F.)
- Department of Cellular Biology, Physiology and Immunology, Campus Bellaterra, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
| | - Silvia Presas-Rodríguez
- Multiple Sclerosis Unit, Department of Neurosciences, Germans Trias i Pujol University Hospital, 08916 Badalona, Spain;
- Department of Medicine, Campus Bellaterra, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
| | - Aina Teniente-Serra
- Immunology Division, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Campus Can Ruti, 08916 Badalona, Spain; (B.Q.-S.); (M.J.M.); (J.N.-B.); (A.T.-S.); (F.F.)
- Department of Cellular Biology, Physiology and Immunology, Campus Bellaterra, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
| | - Federico Fondelli
- Immunology Division, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Campus Can Ruti, 08916 Badalona, Spain; (B.Q.-S.); (M.J.M.); (J.N.-B.); (A.T.-S.); (F.F.)
- Department of Cellular Biology, Physiology and Immunology, Campus Bellaterra, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
| | - Cristina Ramo-Tello
- Multiple Sclerosis Unit, Department of Neurosciences, Germans Trias i Pujol University Hospital, 08916 Badalona, Spain;
- Department of Medicine, Campus Bellaterra, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
- Correspondence: or (C.R.-T.); (E.M.-C.); Tel.: +34-93-497-8433 (C.R.-T.); +34-93-497-8666 (E.M.-C.)
| | - Eva Martínez-Cáceres
- Immunology Division, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Campus Can Ruti, 08916 Badalona, Spain; (B.Q.-S.); (M.J.M.); (J.N.-B.); (A.T.-S.); (F.F.)
- Department of Cellular Biology, Physiology and Immunology, Campus Bellaterra, Universitat Autònoma de Barcelona, 08193 Cerdanyola del Vallès, Spain
- Correspondence: or (C.R.-T.); (E.M.-C.); Tel.: +34-93-497-8433 (C.R.-T.); +34-93-497-8666 (E.M.-C.)
| |
Collapse
|
16
|
Biologia Futura: Emerging antigen-specific therapies for autoimmune diseases. Biol Futur 2021; 72:15-24. [PMID: 34554499 DOI: 10.1007/s42977-021-00074-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 01/16/2021] [Indexed: 02/05/2023]
Abstract
Autoimmune diseases are caused by breaking the central and/or peripheral tolerance against self, leading to uncontrolled immune response to autoantigens. The incidences of autoimmune diseases have increased significantly worldwide over the last decades; nearly 5% of the world's population is affected. The current treatments aim to reduce pain and inflammation to prevent organ damage and have a general immunosuppressive effect, but they cannot cure the disease. There is a huge unmet need for autoantigen-specific therapy, without affecting the immune response against pathogens. This goal can be achieved by targeting autoantigen-specific T or B cells and by restoring self-tolerance by inducing tolerogenic antigen-presenting cells (APC) and the development of regulatory T (Treg) cells, for example, by using autoantigenic peptides bound to nanoparticles. Transferring in vitro manipulated autologous tolerogenic APC or autologous autoantigen-specific Treg cells to patients is the promising approach to develop cellular therapeutics. Most recently, chimeric autoantibody receptor T cells have been designed to specifically deplete autoreactive B cells. Limitations of these novel autoantigen-specific therapies will also be discussed.
Collapse
|
17
|
Passeri L, Marta F, Bassi V, Gregori S. Tolerogenic Dendritic Cell-Based Approaches in Autoimmunity. Int J Mol Sci 2021; 22:8415. [PMID: 34445143 PMCID: PMC8395087 DOI: 10.3390/ijms22168415] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 08/02/2021] [Accepted: 08/03/2021] [Indexed: 12/15/2022] Open
Abstract
Dendritic cells (DCs) dictate the outcomes of tissue-specific immune responses. In the context of autoimmune diseases, DCs instruct T cells to respond to antigens (Ags), including self-Ags, leading to organ damage, or to becoming regulatory T cells (Tregs) promoting and perpetuating immune tolerance. DCs can acquire tolerogenic properties in vitro and in vivo in response to several stimuli, a feature that opens the possibility to generate or to target DCs to restore tolerance in autoimmune settings. We present an overview of the different subsets of human DCs and of the regulatory mechanisms associated with tolerogenic (tol)DC functions. We review the role of DCs in the induction of tissue-specific autoimmunity and the current approaches exploiting tolDC-based therapies or targeting DCs in vivo for the treatment of autoimmune diseases. Finally, we discuss limitations and propose future investigations for improving the knowledge on tolDCs for future clinical assessment to revert and prevent autoimmunity. The continuous expansion of tolDC research areas will lead to improving the understanding of the role that DCs play in the development and treatment of autoimmunity.
Collapse
Affiliation(s)
- Laura Passeri
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (L.P.); (F.M.); (V.B.)
- San Raffaele Scientific Institute IRCCS, University Vita-Salute San Raffaele, 20132 Milan, Italy
| | - Fortunato Marta
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (L.P.); (F.M.); (V.B.)
| | - Virginia Bassi
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (L.P.); (F.M.); (V.B.)
- San Raffaele Scientific Institute IRCCS, University Vita-Salute San Raffaele, 20132 Milan, Italy
| | - Silvia Gregori
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy; (L.P.); (F.M.); (V.B.)
| |
Collapse
|
18
|
Robinson S, Thomas R. Potential for Antigen-Specific Tolerizing Immunotherapy in Systematic Lupus Erythematosus. Front Immunol 2021; 12:654701. [PMID: 34335564 PMCID: PMC8322693 DOI: 10.3389/fimmu.2021.654701] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 06/25/2021] [Indexed: 12/12/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic complex systemic autoimmune disease characterized by multiple autoantibodies and clinical manifestations, with the potential to affect nearly every organ. SLE treatments, including corticosteroids and immunosuppressive drugs, have greatly increased survival rates, but there is no curative therapy and SLE management is limited by drug complications and toxicities. There is an obvious clinical need for safe, effective SLE treatments. A promising treatment avenue is to restore immunological tolerance to reduce inflammatory clinical manifestations of SLE. Indeed, recent clinical trials of low-dose IL-2 supplementation in SLE patients showed that in vivo expansion of regulatory T cells (Treg cells) is associated with dramatic but transient improvement in SLE disease markers and clinical manifestations. However, the Treg cells that expanded were short-lived and unstable. Alternatively, antigen-specific tolerance (ASIT) approaches that establish long-lived immunological tolerance could be deployed in the context of SLE. In this review, we discuss the potential benefits and challenges of nanoparticle ASIT approaches to induce prolonged immunological tolerance in SLE.
Collapse
Affiliation(s)
- Sean Robinson
- School of Medicine, Faculty of Medicine and Biomedical Sciences, University of Queensland, St Lucia, QLD, Australia
| | - Ranjeny Thomas
- University of Queensland Diamantina Institute, The University of Queensland, Princess Alexandra Hospital, Woolloongabba, QLD, Australia
| |
Collapse
|
19
|
Made to Measure: Patient-Tailored Treatment of Multiple Sclerosis Using Cell-Based Therapies. Int J Mol Sci 2021; 22:ijms22147536. [PMID: 34299154 PMCID: PMC8304207 DOI: 10.3390/ijms22147536] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/25/2021] [Accepted: 06/28/2021] [Indexed: 12/14/2022] Open
Abstract
Currently, there is still no cure for multiple sclerosis (MS), which is an autoimmune and neurodegenerative disease of the central nervous system. Treatment options predominantly consist of drugs that affect adaptive immunity and lead to a reduction of the inflammatory disease activity. A broad range of possible cell-based therapeutic options are being explored in the treatment of autoimmune diseases, including MS. This review aims to provide an overview of recent and future advances in the development of cell-based treatment options for the induction of tolerance in MS. Here, we will focus on haematopoietic stem cells, mesenchymal stromal cells, regulatory T cells and dendritic cells. We will also focus on less familiar cell types that are used in cell therapy, including B cells, natural killer cells and peripheral blood mononuclear cells. We will address key issues regarding the depicted therapies and highlight the major challenges that lie ahead to successfully reverse autoimmune diseases, such as MS, while minimising the side effects. Although cell-based therapies are well known and used in the treatment of several cancers, cell-based treatment options hold promise for the future treatment of autoimmune diseases in general, and MS in particular.
Collapse
|
20
|
Mansilla MJ, Presas-Rodríguez S, Teniente-Serra A, González-Larreategui I, Quirant-Sánchez B, Fondelli F, Djedovic N, Iwaszkiewicz-Grześ D, Chwojnicki K, Miljković Đ, Trzonkowski P, Ramo-Tello C, Martínez-Cáceres EM. Paving the way towards an effective treatment for multiple sclerosis: advances in cell therapy. Cell Mol Immunol 2021; 18:1353-1374. [PMID: 33958746 PMCID: PMC8167140 DOI: 10.1038/s41423-020-00618-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 12/03/2020] [Indexed: 12/13/2022] Open
Abstract
Multiple sclerosis (MS) is a leading cause of chronic neurological disability in young to middle-aged adults, affecting ~2.5 million people worldwide. Currently, most therapeutics for MS are systemic immunosuppressive or immunomodulatory drugs, but these drugs are unable to halt or reverse the disease and have the potential to cause serious adverse events. Hence, there is an urgent need for the development of next-generation treatments that, alone or in combination, stop the undesired autoimmune response and contribute to the restoration of homeostasis. This review analyzes current MS treatments as well as different cell-based therapies that have been proposed to restore homeostasis in MS patients (tolerogenic dendritic cells, regulatory T cells, mesenchymal stem cells, and vaccination with T cells). Data collected from preclinical studies performed in the experimental autoimmune encephalomyelitis (EAE) model of MS in animals, in vitro cultures of cells from MS patients and the initial results of phase I/II clinical trials are analyzed to better understand which parameters are relevant for obtaining an efficient cell-based therapy for MS.
Collapse
Affiliation(s)
- M J Mansilla
- Division of Immunology, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Barcelona, Spain. .,Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain.
| | - S Presas-Rodríguez
- Multiple Sclerosis Unit, Department of Neurosciences, Germans Trias i Pujol University Hospital, Barcelona, Spain.,Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - A Teniente-Serra
- Division of Immunology, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Barcelona, Spain.,Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - I González-Larreategui
- Division of Immunology, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Barcelona, Spain.,Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - B Quirant-Sánchez
- Division of Immunology, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Barcelona, Spain.,Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - F Fondelli
- Division of Immunology, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Barcelona, Spain.,Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - N Djedovic
- Department of Immunology, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - D Iwaszkiewicz-Grześ
- Department of Medical Immunology, Medical University of Gdańsk, Gdańsk, Poland.,Poltreg S.A., Gdańsk, Poland
| | - K Chwojnicki
- Department of Anaesthesiology & Intensive Care, Medical University of Gdańsk, Gdańsk, Poland
| | - Đ Miljković
- Department of Immunology, Institute for Biological Research "Siniša Stanković"- National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - P Trzonkowski
- Department of Medical Immunology, Medical University of Gdańsk, Gdańsk, Poland.,Poltreg S.A., Gdańsk, Poland
| | - C Ramo-Tello
- Multiple Sclerosis Unit, Department of Neurosciences, Germans Trias i Pujol University Hospital, Barcelona, Spain.,Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - E M Martínez-Cáceres
- Division of Immunology, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Barcelona, Spain. .,Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain.
| |
Collapse
|
21
|
van Wigcheren GF, Roelofs D, Figdor CG, Flórez-Grau G. Three distinct tolerogenic CD14 + myeloid cell types to actively manage autoimmune disease: Opportunities and challenges. J Autoimmun 2021; 120:102645. [PMID: 33901801 DOI: 10.1016/j.jaut.2021.102645] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/19/2021] [Accepted: 04/19/2021] [Indexed: 01/18/2023]
Abstract
Current treatment for patients with autoimmune disorders including rheumatoid arthritis, multiple sclerosis and type 1 diabetes, often consists of long-term drug regimens that broadly dampen immune responses. These non-specific treatments are frequently associated with severe side effects creating an urgent need for safer and more effective therapy to promote peripheral tolerance in autoimmune diseases. Cell-based immunotherapy may offer an encouraging alternative, where tolerogenic CD14+ myeloid cells are infused to inhibit autoreactive effector cells. In this review, we compared in depth three promising tolerogenic CD14+ candidates for the treatment of autoimmune disease: 1) tolerogenic dendritic cells, 2) monocytic myeloid-derived suppressor cells and 3) CD14+ type 2 conventional dendritic cells. TolDC-based therapy has entered clinical testing whereas evidence from the latter two cell types m-MDSCs and CD14+ cDC2s is predominantly coming from cancer immunology research. These three cell types have distinct cellular properties and immunosuppressive mechanisms offering unique opportunities to be explored. However, these cells differ in stage of development towards immunotherapy each facing additional hurdles. Therefore, we speculate on the potential benefits and risks of these cell types as novel cell-based immunotherapies to control autoimmune disease in patients.
Collapse
Affiliation(s)
- Glenn F van Wigcheren
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, the Netherlands; Oncode Institute, the Netherlands
| | - Daphne Roelofs
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, the Netherlands
| | - Carl G Figdor
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, the Netherlands; Oncode Institute, the Netherlands.
| | - Georgina Flórez-Grau
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, the Netherlands
| |
Collapse
|
22
|
Moorman CD, Sohn SJ, Phee H. Emerging Therapeutics for Immune Tolerance: Tolerogenic Vaccines, T cell Therapy, and IL-2 Therapy. Front Immunol 2021; 12:657768. [PMID: 33854514 PMCID: PMC8039385 DOI: 10.3389/fimmu.2021.657768] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 03/04/2021] [Indexed: 12/14/2022] Open
Abstract
Autoimmune diseases affect roughly 5-10% of the total population, with women affected more than men. The standard treatment for autoimmune or autoinflammatory diseases had long been immunosuppressive agents until the advent of immunomodulatory biologic drugs, which aimed at blocking inflammatory mediators, including proinflammatory cytokines. At the frontier of these biologic drugs are TNF-α blockers. These therapies inhibit the proinflammatory action of TNF-α in common autoimmune diseases such as rheumatoid arthritis, psoriasis, ulcerative colitis, and Crohn's disease. TNF-α blockade quickly became the "standard of care" for these autoimmune diseases due to their effectiveness in controlling disease and decreasing patient's adverse risk profiles compared to broad-spectrum immunosuppressive agents. However, anti-TNF-α therapies have limitations, including known adverse safety risk, loss of therapeutic efficacy due to drug resistance, and lack of efficacy in numerous autoimmune diseases, including multiple sclerosis. The next wave of truly transformative therapeutics should aspire to provide a cure by selectively suppressing pathogenic autoantigen-specific immune responses while leaving the rest of the immune system intact to control infectious diseases and malignancies. In this review, we will focus on three main areas of active research in immune tolerance. First, tolerogenic vaccines aiming at robust, lasting autoantigen-specific immune tolerance. Second, T cell therapies using Tregs (either polyclonal, antigen-specific, or genetically engineered to express chimeric antigen receptors) to establish active dominant immune tolerance or T cells (engineered to express chimeric antigen receptors) to delete pathogenic immune cells. Third, IL-2 therapies aiming at expanding immunosuppressive regulatory T cells in vivo.
Collapse
Affiliation(s)
| | | | - Hyewon Phee
- Department of Inflammation and Oncology, Amgen Research, Amgen Inc., South San Francisco, CA, United States
| |
Collapse
|
23
|
Derdelinckx J, Cras P, Berneman ZN, Cools N. Antigen-Specific Treatment Modalities in MS: The Past, the Present, and the Future. Front Immunol 2021; 12:624685. [PMID: 33679769 PMCID: PMC7933447 DOI: 10.3389/fimmu.2021.624685] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 01/04/2021] [Indexed: 12/15/2022] Open
Abstract
Antigen-specific therapy for multiple sclerosis may lead to a more effective therapy by induction of tolerance to a wide range of myelin-derived antigens without hampering the normal surveillance and effector function of the immune system. Numerous attempts to restore tolerance toward myelin-derived antigens have been made over the past decades, both in animal models of multiple sclerosis and in clinical trials for multiple sclerosis patients. In this review, we will give an overview of the current approaches for antigen-specific therapy that are in clinical development for multiple sclerosis as well provide an insight into the challenges for future antigen-specific treatment strategies for multiple sclerosis.
Collapse
Affiliation(s)
- Judith Derdelinckx
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VaxInfectio), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.,Division of Neurology, Antwerp University Hospital, Edegem, Belgium
| | - Patrick Cras
- Division of Neurology, Antwerp University Hospital, Edegem, Belgium.,Born Bunge Institute, Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Zwi N Berneman
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VaxInfectio), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.,Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Nathalie Cools
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (VaxInfectio), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium.,Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium
| |
Collapse
|
24
|
Navarro-Barriuso J, Mansilla MJ, Quirant-Sánchez B, Teniente-Serra A, Ramo-Tello C, Martínez-Cáceres EM. Vitamin D3-Induced Tolerogenic Dendritic Cells Modulate the Transcriptomic Profile of T CD4 + Cells Towards a Functional Hyporesponsiveness. Front Immunol 2021; 11:599623. [PMID: 33552054 PMCID: PMC7856150 DOI: 10.3389/fimmu.2020.599623] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 11/30/2020] [Indexed: 12/22/2022] Open
Abstract
The use of autologous tolerogenic dendritic cells (tolDC) has become a promising alternative for the treatment of autoimmune diseases. Among the different strategies available, the use of vitamin D3 for the generation of tolDC (vitD3-tolDC) constitutes one of the most robust approaches due to their immune regulatory properties, which are currently being tested in clinical trials. However, the mechanisms that vitD3-tolDC trigger for the induction of tolerance remain elusive. For this reason, we performed a full phenotypical, functional, and transcriptomic characterization of T cells upon their interaction with autologous, antigen-specific vitD3-tolDC. We observed a strong antigen-specific reduction of T cell proliferation, combined with a decrease in the relative prevalence of TH1 subpopulations and IFN-γ production. The analysis of the transcriptomic profile of T CD4+ cells evidenced a significant down-modulation of genes involved in cell cycle and cell response to mainly pro-inflammatory immune-related stimuli, highlighting the role of JUNB gene as a potential biomarker of these processes. Consequently, our results show the induction of a strong antigen-specific hyporesponsiveness combined with a reduction on the TH1 immune profile of T cells upon their interaction with vitD3-tolDC, which manifests the regulatory properties of these cells and, therefore, their therapeutic potential in the clinic.
Collapse
Affiliation(s)
- Juan Navarro-Barriuso
- Division of Immunology, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Barcelona, Spain
- Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - María José Mansilla
- Division of Immunology, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Barcelona, Spain
- Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Bibiana Quirant-Sánchez
- Division of Immunology, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Barcelona, Spain
- Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Aina Teniente-Serra
- Division of Immunology, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Barcelona, Spain
- Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Cristina Ramo-Tello
- Multiple Sclerosis Unit, Department of Neurosciences, Germans Trias i Pujol University Hospital, Barcelona, Spain
| | - Eva M. Martínez-Cáceres
- Division of Immunology, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Barcelona, Spain
- Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
25
|
Morante-Palacios O, Fondelli F, Ballestar E, Martínez-Cáceres EM. Tolerogenic Dendritic Cells in Autoimmunity and Inflammatory Diseases. Trends Immunol 2020; 42:59-75. [PMID: 33293219 DOI: 10.1016/j.it.2020.11.001] [Citation(s) in RCA: 158] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 11/06/2020] [Accepted: 11/08/2020] [Indexed: 12/11/2022]
Abstract
Dendritic cells (DCs), the most efficient antigen-presenting cells, are necessary for the effective activation of naïve T cells. DCs can also acquire tolerogenic functions in vivo and in vitro in response to various stimuli, including interleukin (IL)-10, transforming growth factor (TGF)-β, vitamin D3, corticosteroids, and rapamycin. In this review, we provide a wide perspective on the regulatory mechanisms, including crosstalk with other cell types, downstream signaling pathways, transcription factors, and epigenetics, underlying the acquisition of tolerogenesis by DCs, with a special focus on human studies. Finally, we present clinical assays targeting, or based on, tolerogenic DCs in inflammatory diseases. Our discussion provides a useful resource for better understanding the biology of tolerogenic DCs and their manipulation to improve the immunological fitness of patients with certain inflammatory conditions.
Collapse
Affiliation(s)
- Octavio Morante-Palacios
- Epigenetics and Immune Disease Group, Josep Carreras Research Institute (IJC), 08916 Badalona, Barcelona, Spain; Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Barcelona, Spain
| | - Federico Fondelli
- Division of Immunology, Germans Trias i Pujol Hospital, LCMN, Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Barcelona, Spain; Department of Cell Biology, Physiology, Immunology, Autonomous University of Barcelona, 08193 Bellaterra, Barcelona, Spain
| | - Esteban Ballestar
- Epigenetics and Immune Disease Group, Josep Carreras Research Institute (IJC), 08916 Badalona, Barcelona, Spain; Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Barcelona, Spain.
| | - Eva M Martínez-Cáceres
- Division of Immunology, Germans Trias i Pujol Hospital, LCMN, Germans Trias i Pujol Research Institute (IGTP), 08916 Badalona, Barcelona, Spain; Department of Cell Biology, Physiology, Immunology, Autonomous University of Barcelona, 08193 Bellaterra, Barcelona, Spain.
| |
Collapse
|
26
|
Christofi M, Le Sommer S, Mölzer C, Klaska IP, Kuffova L, Forrester JV. Low-dose 2-deoxy glucose stabilises tolerogenic dendritic cells and generates potent in vivo immunosuppressive effects. Cell Mol Life Sci 2020; 78:2857-2876. [PMID: 33074350 PMCID: PMC8004500 DOI: 10.1007/s00018-020-03672-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 09/04/2020] [Accepted: 10/05/2020] [Indexed: 01/17/2023]
Abstract
Cell therapies for autoimmune diseases using tolerogenic dendritic cells (tolDC) have been promisingly explored. A major stumbling block has been generating stable tolDC, with low risk of converting to mature immunogenic DC (mDC), exacerbating disease. mDC induction involves a metabolic shift to lactate production from oxidative phosphorylation (OXPHOS) and β-oxidation, the homeostatic energy source for resting DC. Inhibition of glycolysis through the administration of 2-deoxy glucose (2-DG) has been shown to prevent autoimmune disease experimentally but is not clinically feasible. We show here that treatment of mouse bone marrow-derived tolDC ex vivo with low-dose 2-DG (2.5 mM) (2-DGtolDC) induces a stable tolerogenic phenotype demonstrated by their failure to engage lactate production when challenged with mycobacterial antigen (Mtb). ~ 15% of 2-DGtolDC express low levels of MHC class II and 30% express CD86, while they are negative for CD40. 2-DGtolDC also express increased immune checkpoint molecules PDL-1 and SIRP-1α. Antigen-specific T cell proliferation is reduced in response to 2-DGtolDC in vitro. Mtb-stimulated 2-DGtolDC do not engage aerobic glycolysis but respond to challenge via increased OXPHOS. They also have decreased levels of p65 phosphorylation, with increased phosphorylation of the non-canonical p100 pathway. A stable tolDC phenotype is associated with sustained SIRP-1α phosphorylation and p85-AKT and PI3K signalling inhibition. Further, 2-DGtolDC preferentially secrete IL-10 rather than IL-12 upon Mtb-stimulation. Importantly, a single subcutaneous administration of 2-DGtolDC prevented experimental autoimmune uveoretinitis (EAU) in vivo. Inhibiting glycolysis of autologous tolDC prior to transfer may be a useful approach to providing stable tolDC therapy for autoimmune/immune-mediated diseases.
Collapse
Affiliation(s)
- M Christofi
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, Foresterhill, University of Aberdeen, Aberdeen, AB25 2ZD, Scotland, UK
| | - S Le Sommer
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, Foresterhill, University of Aberdeen, Aberdeen, AB25 2ZD, Scotland, UK
| | - C Mölzer
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, Foresterhill, University of Aberdeen, Aberdeen, AB25 2ZD, Scotland, UK.
| | - I P Klaska
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, Foresterhill, University of Aberdeen, Aberdeen, AB25 2ZD, Scotland, UK
| | - L Kuffova
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, Foresterhill, University of Aberdeen, Aberdeen, AB25 2ZD, Scotland, UK.,Eye Clinic, Aberdeen Royal Infirmary, Aberdeen, Scotland, UK
| | - J V Forrester
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, Foresterhill, University of Aberdeen, Aberdeen, AB25 2ZD, Scotland, UK. .,Ocular Immunology Program, Centre for Ophthalmology and Visual Science, The University of Western Australia, Perth, WA, Australia. .,Centre for Experimental Immunology, Lions Eye Institute, Nedlands, WA, Australia.
| |
Collapse
|
27
|
Dendritic Cells Transfected with MHC Antigenic Determinants of CBA Mice Induce Antigen-Specific Tolerance in C57Bl/6 Mice. J Immunol Res 2020; 2020:9686143. [PMID: 32953894 PMCID: PMC7487104 DOI: 10.1155/2020/9686143] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 07/07/2020] [Accepted: 07/31/2020] [Indexed: 12/12/2022] Open
Abstract
Background Nonspecific immunosuppressive therapy for graft rejection and graft-versus-host disease (GVHD) is often accompanied by severe side effects such as opportunistic infections and cancers. Several approaches have been developed to suppress transplantation reactions using tolerogenic cells, including induction of FoxP3+ Tregs with antigen-loaded dendritic cells (DCs) and induction of CD4+IL-10+ cells with interleukin IL-10-producing DCs. Here, we assessed the effectiveness of both approaches in the suppression of graft rejection and GVHD. Methods IL-10-producing DCs were generated by the transfection of DCs with DNA constructs encoding mouse IL-10. Antigen-loaded DCs from C57BL/6 mice were generated by transfection with DNA constructs encoding antigenic determinants from the H2 locus of CBA mice which differ from the homologous antigenic determinants of C57BL/6 mice. Results We found that both IL-10-producing DCs and antigen-loaded immature DCs could suppress graft rejection and GVHD but through distinct nonspecific and antigen-specific mechanisms, respectively. Discussion. We provide data that the novel approach for DCs antigen loading using DNA constructs encoding distinct homologous determinants derived from major histocompatibility complex genes is effective in antigen-specific suppression of transplantation reactions. Such an approach eliminates the necessity of donor material use and may be useful in immunosuppressive therapy side effects prevention.
Collapse
|
28
|
Recent Advances in Antigen-Specific Immunotherapies for the Treatment of Multiple Sclerosis. Brain Sci 2020; 10:brainsci10060333. [PMID: 32486045 PMCID: PMC7348736 DOI: 10.3390/brainsci10060333] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 05/22/2020] [Accepted: 05/26/2020] [Indexed: 12/11/2022] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disease of the central nervous system and is considered to be the leading non-traumatic cause of neurological disability in young adults. Current treatments for MS comprise long-term immunosuppressant drugs and disease-modifying therapies (DMTs) designed to alter its progress with the enhanced risk of severe side effects. The Holy Grail for the treatment of MS is to specifically suppress the disease while at the same time allow the immune system to be functionally active against infectious diseases and malignancy. This could be achieved via the development of immunotherapies designed to specifically suppress immune responses to self-antigens (e.g., myelin antigens). The present study attempts to highlight the various antigen-specific immunotherapies developed so far for the treatment of multiple sclerosis (e.g., vaccination with myelin-derived peptides/proteins, plasmid DNA encoding myelin epitopes, tolerogenic dendritic cells pulsed with encephalitogenic epitopes of myelin proteins, attenuated autologous T cells specific for myelin antigens, T cell receptor peptides, carriers loaded/conjugated with myelin immunodominant peptides, etc), focusing on the outcome of their recent preclinical and clinical evaluation, and to shed light on the mechanisms involved in the immunopathogenesis and treatment of multiple sclerosis.
Collapse
|
29
|
Metaxakis A, Petratou D, Tavernarakis N. Molecular Interventions towards Multiple Sclerosis Treatment. Brain Sci 2020; 10:brainsci10050299. [PMID: 32429225 PMCID: PMC7287961 DOI: 10.3390/brainsci10050299] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 05/11/2020] [Accepted: 05/12/2020] [Indexed: 12/24/2022] Open
Abstract
Multiple sclerosis (MS) is an autoimmune life-threatening disease, afflicting millions of people worldwide. Although the disease is non-curable, considerable therapeutic advances have been achieved through molecular immunotherapeutic approaches, such as peptides vaccination, administration of monoclonal antibodies, and immunogenic copolymers. The main aims of these therapeutic strategies are to shift the MS-related autoimmune response towards a non-inflammatory T helper 2 (Th2) cells response, inactivate or ameliorate cytotoxic autoreactive T cells, induce secretion of anti-inflammatory cytokines, and inhibit recruitment of autoreactive lymphocytes to the central nervous system (CNS). These approaches can efficiently treat autoimmune encephalomyelitis (EAE), an essential system to study MS in animals, but they can only partially inhibit disease progress in humans. Nevertheless, modern immunotherapeutic techniques remain the most promising tools for the development of safe MS treatments, specifically targeting the cellular factors that trigger the initiation of the disease.
Collapse
Affiliation(s)
- Athanasios Metaxakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Nikolaou Plastira 100, 70013 Heraklion, Greece; (A.M.); (D.P.)
| | - Dionysia Petratou
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Nikolaou Plastira 100, 70013 Heraklion, Greece; (A.M.); (D.P.)
| | - Nektarios Tavernarakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Nikolaou Plastira 100, 70013 Heraklion, Greece; (A.M.); (D.P.)
- Department of Basic Sciences, Faculty of Medicine, University of Crete, 71110 Heraklion, Greece
- Correspondence: ; Tel.: +30-2810-391066
| |
Collapse
|
30
|
Marin E, Bouchet-Delbos L, Renoult O, Louvet C, Nerriere-Daguin V, Managh AJ, Even A, Giraud M, Vu Manh TP, Aguesse A, Bériou G, Chiffoleau E, Alliot-Licht B, Prieur X, Croyal M, Hutchinson JA, Obermajer N, Geissler EK, Vanhove B, Blancho G, Dalod M, Josien R, Pecqueur C, Cuturi MC, Moreau A. Human Tolerogenic Dendritic Cells Regulate Immune Responses through Lactate Synthesis. Cell Metab 2019; 30:1075-1090.e8. [PMID: 31801055 DOI: 10.1016/j.cmet.2019.11.011] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 07/17/2019] [Accepted: 11/12/2019] [Indexed: 12/22/2022]
Abstract
Cell therapy is a promising strategy for treating patients suffering from autoimmune or inflammatory diseases or receiving a transplant. Based on our preclinical studies, we have generated human autologous tolerogenic dendritic cells (ATDCs), which are being tested in a first-in-man clinical trial in kidney transplant recipients. Here, we report that ATDCs represent a unique subset of monocyte-derived cells based on phenotypic, transcriptomic, and metabolic analyses. ATDCs are characterized by their suppression of T cell proliferation and their expansion of Tregs through secreted factors. ATDCs produce high levels of lactate that shape T cell responses toward tolerance. Indeed, T cells take up ATDC-secreted lactate, leading to a decrease of their glycolysis. In vivo, ATDCs promote elevated levels of circulating lactate and delay graft-versus-host disease by reducing T cell proliferative capacity. The suppression of T cell immunity through lactate production by ATDCs is a novel mechanism that distinguishes ATDCs from other cell-based immunotherapies.
Collapse
Affiliation(s)
- Eros Marin
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, CHU Nantes, ITUN, Nantes, France
| | - Laurence Bouchet-Delbos
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, CHU Nantes, ITUN, Nantes, France
| | - Ophélie Renoult
- Centre de Recherche en Cancérologie et Immunologie Nantes-Angers UMR1232, INSERM, Université de Nantes, Nantes, France
| | - Cédric Louvet
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, CHU Nantes, ITUN, Nantes, France
| | - Véronique Nerriere-Daguin
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, CHU Nantes, ITUN, Nantes, France; LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France
| | - Amy J Managh
- Centre for Analytical Science, Department of Chemistry, Loughborough University, Loughborough, UK
| | - Amandine Even
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, CHU Nantes, ITUN, Nantes, France
| | - Matthieu Giraud
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, CHU Nantes, ITUN, Nantes, France
| | - Thien Phong Vu Manh
- Centre d'Immunologie de Marseille-Luminy, Aix-Marseille Université, CNRS, INSERM, CIML, Marseille, France
| | - Audrey Aguesse
- UMR 1280 PhAN, Mass Spectrometry Core Facility, INRA, CRNHO, West Human Nutrition Research Center, Nantes, France
| | - Gaelle Bériou
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, CHU Nantes, ITUN, Nantes, France
| | - Elise Chiffoleau
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, CHU Nantes, ITUN, Nantes, France
| | - Brigitte Alliot-Licht
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, CHU Nantes, ITUN, Nantes, France; Faculté d'Odontologie, Université de Nantes, Nantes, France
| | - Xavier Prieur
- Institut du Thorax, INSERM, CNRS, Université de Nantes, Nantes, France
| | - Mikael Croyal
- UMR 1280 PhAN, Mass Spectrometry Core Facility, INRA, CRNHO, West Human Nutrition Research Center, Nantes, France
| | - James A Hutchinson
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Natasa Obermajer
- Division of Surgical Oncology, University of Pittsburgh, Hillman Cancer Center, Pittsburgh, PA, USA
| | - Edward K Geissler
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Bernard Vanhove
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, CHU Nantes, ITUN, Nantes, France; LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France
| | - Gilles Blancho
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, CHU Nantes, ITUN, Nantes, France
| | - Marc Dalod
- Centre d'Immunologie de Marseille-Luminy, Aix-Marseille Université, CNRS, INSERM, CIML, Marseille, France
| | - Régis Josien
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, CHU Nantes, ITUN, Nantes, France; Laboratoire d'Immunologie, CHU Nantes, Nantes Université, Nantes, France
| | - Claire Pecqueur
- Centre de Recherche en Cancérologie et Immunologie Nantes-Angers UMR1232, INSERM, Université de Nantes, Nantes, France; LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France
| | - Maria-Cristina Cuturi
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, CHU Nantes, ITUN, Nantes, France; LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France
| | - Aurélie Moreau
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, CHU Nantes, ITUN, Nantes, France; LabEx IGO "Immunotherapy, Graft, Oncology", Nantes, France.
| |
Collapse
|
31
|
Funes SC, Ríos M, Gómez‐Santander F, Fernández‐Fierro A, Altamirano‐Lagos MJ, Rivera‐Perez D, Pulgar‐Sepúlveda R, Jara EL, Rebolledo‐Zelada D, Villarroel A, Roa JC, Mackern‐Oberti JP, Kalergis AM. Tolerogenic dendritic cell transfer ameliorates systemic lupus erythematosus in mice. Immunology 2019; 158:322-339. [PMID: 31509246 PMCID: PMC6856940 DOI: 10.1111/imm.13119] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 08/28/2019] [Accepted: 09/05/2019] [Indexed: 12/11/2022] Open
Abstract
Current treatments for systemic autoimmune diseases partially improve the health of patients displaying low pharmacological efficacy and systemic immunosuppression. Here, the therapeutic potential of transferring tolerogenic dendritic cells (tolDCs) generated with heme-oxygenase inductor cobalt (III) protoporphyrin IX (CoPP), dexamethasone and rosiglitazone for the treatment of systemic autoimmunity was evaluated in two murine models of systemic lupus erythematosus (SLE), MRL-Faslpr and NZM2410 mice. Dendritic cells treated ex vivo with these drugs showed a stable tolerogenic profile after lipopolysaccharide stimulation. Regular doses of tolDCs were administered to anti-nuclear antibody-positive mice throughout 60-70 days, and the clinical score was evaluated. Long-term treatment with these tolDCs was well tolerated and effective to improve the clinical score on MRL-Faslpr lupus-prone mice. Additionally, decreased levels of anti-nuclear antibodies in NZM2410 mice were observed. Although tolDC treatment increased regulatory T cells, no significant reduction of renal damage or glomerulonephritis could be found. In conclusion, these results suggest that the transfer of histone-loaded tolDCs could improve only some SLE symptoms and reduced anti-nuclear antibodies. This is the first study to evaluate antigen-specific tolDC administration to treat SLE. Our report strengthens the clinical relevance of tolDC generation with CoPP, dexamethasone and rosiglitazone and the use of these modified cells as a therapy for systemic autoimmunity.
Collapse
Affiliation(s)
- Samanta C. Funes
- Millennium Institute on Immunology and ImmunotherapyDepartamento de Genética Molecular y MicrobiologíaFacultad de Ciencias BiológicasPontificia Universidad Católica de ChileSantiagoChile
| | - Mariana Ríos
- Millennium Institute on Immunology and ImmunotherapyDepartamento de Genética Molecular y MicrobiologíaFacultad de Ciencias BiológicasPontificia Universidad Católica de ChileSantiagoChile
| | - Felipe Gómez‐Santander
- Millennium Institute on Immunology and ImmunotherapyDepartamento de Genética Molecular y MicrobiologíaFacultad de Ciencias BiológicasPontificia Universidad Católica de ChileSantiagoChile
| | - Ayleen Fernández‐Fierro
- Millennium Institute on Immunology and ImmunotherapyDepartamento de Genética Molecular y MicrobiologíaFacultad de Ciencias BiológicasPontificia Universidad Católica de ChileSantiagoChile
| | - María J. Altamirano‐Lagos
- Millennium Institute on Immunology and ImmunotherapyDepartamento de Genética Molecular y MicrobiologíaFacultad de Ciencias BiológicasPontificia Universidad Católica de ChileSantiagoChile
| | - Daniela Rivera‐Perez
- Millennium Institute on Immunology and ImmunotherapyDepartamento de Genética Molecular y MicrobiologíaFacultad de Ciencias BiológicasPontificia Universidad Católica de ChileSantiagoChile
| | - Raul Pulgar‐Sepúlveda
- Millennium Institute on Immunology and ImmunotherapyDepartamento de Genética Molecular y MicrobiologíaFacultad de Ciencias BiológicasPontificia Universidad Católica de ChileSantiagoChile
| | - Evelyn L. Jara
- Millennium Institute on Immunology and ImmunotherapyDepartamento de Genética Molecular y MicrobiologíaFacultad de Ciencias BiológicasPontificia Universidad Católica de ChileSantiagoChile
- Present address:
Facultad de CienciasDepartamento de Ciencias BásicasUniversidad Santo TomásTemucoChile
| | - Diego Rebolledo‐Zelada
- Millennium Institute on Immunology and ImmunotherapyDepartamento de Genética Molecular y MicrobiologíaFacultad de Ciencias BiológicasPontificia Universidad Católica de ChileSantiagoChile
| | - Alejandra Villarroel
- Departamento de Anatomía PatológicaFacultad de MedicinaPontificia Universidad Católica de ChileSantiagoChile
| | - Juan C. Roa
- Departamento de Anatomía PatológicaFacultad de MedicinaPontificia Universidad Católica de ChileSantiagoChile
| | - Juan P. Mackern‐Oberti
- Instituto de Medicina y Biología Experimental de CuyoIMBECU CCT Mendoza‐ CONICETMendozaArgentina
- Instituto de FisiologíaFacultad de Ciencias MédicasUniversidad Nacional de CuyoMendozaArgentina
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and ImmunotherapyDepartamento de Genética Molecular y MicrobiologíaFacultad de Ciencias BiológicasPontificia Universidad Católica de ChileSantiagoChile
- Departamento de EndocrinologíaEscuela de Medicina, Facultad de MedicinaPontificia Universidad Católica de ChileSantiagoChile
| |
Collapse
|
32
|
Spiering R, Jansen MAA, Wood MJ, Fath AA, Eltherington O, Anderson AE, Pratt AG, van Eden W, Isaacs JD, Broere F, Hilkens CMU. Targeting of tolerogenic dendritic cells to heat-shock proteins in inflammatory arthritis. J Transl Med 2019; 17:375. [PMID: 31727095 PMCID: PMC6857208 DOI: 10.1186/s12967-019-2128-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 11/05/2019] [Indexed: 12/21/2022] Open
Abstract
Background Autologous tolerogenic dendritic cells (tolDC) are a promising therapeutic strategy for inflammatory arthritis (IA) as they can regulate autoantigen-specific T cell responses. Here, we investigated two outstanding priorities for clinical development: (i) the suitability of using heat-shock proteins (HSP), abundant in inflamed synovia, as surrogate autoantigens to be presented by tolDC and (ii) identification of functional biomarkers that confirm tolDC regulatory activity. Methods Cell proliferation dye-labelled human peripheral blood mononuclear cells of IA (rheumatoid arthritis (RA) and psoriatic arthritis (PsA)) patients or healthy donors were cultured with HSP40-, HSP60- and HSP70-derived peptides or recall antigens (e.g. tuberculin purified protein derivative (PPD)) in the presence or absence of tolDC or control DC for 9 days. Functional characteristics of proliferated antigen-specific T-cells were measured using flow cytometry, gene expression profiling and cytokine secretion immunoassays. Repeated measures analysis of variance (ANOVA) with Bonferroni correction for comparisons between multiple groups and paired Student t test for comparisons between two groups were used to determine significance. Results All groups showed robust CD4+ T-cell responses towards one or more HSP-derived peptide(s) as assessed by a stimulation index > 2 (healthy donors: 78%, RA: 73%, PsA: 90%) and production of the cytokines IFNγ, IL-17A and GM-CSF. Addition of tolDC but not control DC induced a type 1 regulatory (Tr1) phenotype in the antigen-specific CD4+ T-cell population, as identified by high expression of LAG3, CD49b and secretion of IL-10. Furthermore, tolDC inhibited bystander natural killer (NK) cell activation in a TGFβ dependent manner. Conclusions HSP-specific CD4+ T-cells are detectable in the majority of RA and PsA patients and can be converted into Tr1 cells by tolDC. HSP-loaded tolDC may therefore be suitable for directing T regulatory responses to antigens in inflamed synovia of IA patients. Tr1 markers LAG3, CD49b and IL-10 are suitable biomarkers for future tolDC clinical trials.
Collapse
Affiliation(s)
- Rachel Spiering
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK.,Research into Inflammatory Arthritis Centre Versus Arthritis, (Formerly: Arthritis Research UK Rheumatoid Arthritis Pathogenesis Centre of Excellence (RACE)), Newcastle upon Tyne, UK.,NIHR-Newcastle Biomedical Research Centre in Ageing and Long-Term Conditions, Newcastle Upon Tyne Hospitals NHS Foundation Trust and Newcastle University, Newcastle upon Tyne, UK
| | - Manon A A Jansen
- Division of Immunology, Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, The Netherlands
| | - Matthew J Wood
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK.,Research into Inflammatory Arthritis Centre Versus Arthritis, (Formerly: Arthritis Research UK Rheumatoid Arthritis Pathogenesis Centre of Excellence (RACE)), Newcastle upon Tyne, UK.,NIHR-Newcastle Biomedical Research Centre in Ageing and Long-Term Conditions, Newcastle Upon Tyne Hospitals NHS Foundation Trust and Newcastle University, Newcastle upon Tyne, UK
| | - Anshorulloh A Fath
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK.,Research into Inflammatory Arthritis Centre Versus Arthritis, (Formerly: Arthritis Research UK Rheumatoid Arthritis Pathogenesis Centre of Excellence (RACE)), Newcastle upon Tyne, UK.,NIHR-Newcastle Biomedical Research Centre in Ageing and Long-Term Conditions, Newcastle Upon Tyne Hospitals NHS Foundation Trust and Newcastle University, Newcastle upon Tyne, UK
| | - Oliver Eltherington
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK.,Research into Inflammatory Arthritis Centre Versus Arthritis, (Formerly: Arthritis Research UK Rheumatoid Arthritis Pathogenesis Centre of Excellence (RACE)), Newcastle upon Tyne, UK.,NIHR-Newcastle Biomedical Research Centre in Ageing and Long-Term Conditions, Newcastle Upon Tyne Hospitals NHS Foundation Trust and Newcastle University, Newcastle upon Tyne, UK
| | - Amy E Anderson
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK.,Research into Inflammatory Arthritis Centre Versus Arthritis, (Formerly: Arthritis Research UK Rheumatoid Arthritis Pathogenesis Centre of Excellence (RACE)), Newcastle upon Tyne, UK.,NIHR-Newcastle Biomedical Research Centre in Ageing and Long-Term Conditions, Newcastle Upon Tyne Hospitals NHS Foundation Trust and Newcastle University, Newcastle upon Tyne, UK
| | - Arthur G Pratt
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK.,Research into Inflammatory Arthritis Centre Versus Arthritis, (Formerly: Arthritis Research UK Rheumatoid Arthritis Pathogenesis Centre of Excellence (RACE)), Newcastle upon Tyne, UK.,NIHR-Newcastle Biomedical Research Centre in Ageing and Long-Term Conditions, Newcastle Upon Tyne Hospitals NHS Foundation Trust and Newcastle University, Newcastle upon Tyne, UK
| | - Willem van Eden
- Division of Immunology, Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, The Netherlands
| | - John D Isaacs
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK.,Research into Inflammatory Arthritis Centre Versus Arthritis, (Formerly: Arthritis Research UK Rheumatoid Arthritis Pathogenesis Centre of Excellence (RACE)), Newcastle upon Tyne, UK.,NIHR-Newcastle Biomedical Research Centre in Ageing and Long-Term Conditions, Newcastle Upon Tyne Hospitals NHS Foundation Trust and Newcastle University, Newcastle upon Tyne, UK
| | - Femke Broere
- Division of Immunology, Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, The Netherlands.,Department of Clinical Sciences of Companion Animals, Faculty Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Catharien M U Hilkens
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK. .,Research into Inflammatory Arthritis Centre Versus Arthritis, (Formerly: Arthritis Research UK Rheumatoid Arthritis Pathogenesis Centre of Excellence (RACE)), Newcastle upon Tyne, UK. .,NIHR-Newcastle Biomedical Research Centre in Ageing and Long-Term Conditions, Newcastle Upon Tyne Hospitals NHS Foundation Trust and Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
33
|
Willekens B, Presas-Rodríguez S, Mansilla MJ, Derdelinckx J, Lee WP, Nijs G, De Laere M, Wens I, Cras P, Parizel P, Van Hecke W, Ribbens A, Billiet T, Adams G, Couttenye MM, Navarro-Barriuso J, Teniente-Serra A, Quirant-Sánchez B, Lopez-Diaz de Cerio A, Inogés S, Prosper F, Kip A, Verheij H, Gross CC, Wiendl H, Van Ham MS, Ten Brinke A, Barriocanal AM, Massuet-Vilamajó A, Hens N, Berneman Z, Martínez-Cáceres E, Cools N, Ramo-Tello C. Tolerogenic dendritic cell-based treatment for multiple sclerosis (MS): a harmonised study protocol for two phase I clinical trials comparing intradermal and intranodal cell administration. BMJ Open 2019; 9:e030309. [PMID: 31501122 PMCID: PMC6738722 DOI: 10.1136/bmjopen-2019-030309] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
INTRODUCTION Based on the advances in the treatment of multiple sclerosis (MS), currently available disease-modifying treatments (DMT) have positively influenced the disease course of MS. However, the efficacy of DMT is highly variable and increasing treatment efficacy comes with a more severe risk profile. Hence, the unmet need for safer and more selective treatments remains. Specifically restoring immune tolerance towards myelin antigens may provide an attractive alternative. In this respect, antigen-specific tolerisation with autologous tolerogenic dendritic cells (tolDC) is a promising approach. METHODS AND ANALYSIS Here, we will evaluate the clinical use of tolDC in a well-defined population of MS patients in two phase I clinical trials. In doing so, we aim to compare two ways of tolDC administration, namely intradermal and intranodal. The cells will be injected at consecutive intervals in three cohorts receiving incremental doses of tolDC, according to a best-of-five design. The primary objective is to assess the safety and feasibility of tolDC administration. For safety, the number of adverse events including MRI and clinical outcomes will be assessed. For feasibility, successful production of tolDC will be determined. Secondary endpoints include clinical and MRI outcome measures. The patients' immune profile will be assessed to find presumptive evidence for a tolerogenic effect in vivo. ETHICS AND DISSEMINATION Ethics approval was obtained for the two phase I clinical trials. The results of the trials will be disseminated in a peer-reviewed journal, at scientific conferences and to patient associations. TRIAL REGISTRATION NUMBERS NCT02618902 and NCT02903537; EudraCT numbers: 2015-002975-16 and 2015-003541-26.
Collapse
Affiliation(s)
- Barbara Willekens
- Department of Neurology, University Hospital Antwerp, Edegem, Belgium
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp Faculty of Medicine and Health Sciences, Wilrijk, Belgium
| | - Silvia Presas-Rodríguez
- Multiple Sclerosis Unit, Department of Neurosciences, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Cerdanyola del Vallés, Spain
| | - M J Mansilla
- Division of Immunology, LCMN, Hospital Universitario Germans Trias i Pujol and Research Institute, Badalona, Spain
- Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Judith Derdelinckx
- Department of Neurology, University Hospital Antwerp, Edegem, Belgium
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp Faculty of Medicine and Health Sciences, Wilrijk, Belgium
| | - Wai-Ping Lee
- Center for Cell Therapy and Regenerative Medicine, University Hospital Antwerp, Edegem, Belgium
| | - Griet Nijs
- Center for Cell Therapy and Regenerative Medicine, University Hospital Antwerp, Edegem, Belgium
| | - Maxime De Laere
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp Faculty of Medicine and Health Sciences, Wilrijk, Belgium
| | - Inez Wens
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp Faculty of Medicine and Health Sciences, Wilrijk, Belgium
| | - Patrick Cras
- Department of Neurology, University Hospital Antwerp, Edegem, Belgium
| | - Paul Parizel
- Department of Radiology, University Hospital Antwerp, Edegem, Belgium
| | | | | | | | | | | | - Juan Navarro-Barriuso
- Division of Immunology, LCMN, Hospital Universitario Germans Trias i Pujol and Research Institute, Badalona, Spain
- Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Aina Teniente-Serra
- Division of Immunology, LCMN, Hospital Universitario Germans Trias i Pujol and Research Institute, Badalona, Spain
- Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Bibiana Quirant-Sánchez
- Division of Immunology, LCMN, Hospital Universitario Germans Trias i Pujol and Research Institute, Badalona, Spain
- Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Ascensión Lopez-Diaz de Cerio
- Haematology-Cell Therapy Area, clinica universidad de navarra, Pamplona, Spain
- Immunology and Immunotherapy Department, Clinica Universidad de Navarra, Pamplona, Spain
| | - Susana Inogés
- Haematology-Cell Therapy Area, clinica universidad de navarra, Pamplona, Spain
- Immunology and Immunotherapy Department, Clinica Universidad de Navarra, Pamplona, Spain
| | - Felipe Prosper
- Haematology-Cell Therapy Area, clinica universidad de navarra, Pamplona, Spain
- Program of Haematology-Oncology, CIMA, Universidad de Navarra, Pamplona, Spain
| | - Anke Kip
- Lygature, Utrecht, The Netherlands
| | | | - Catharina C Gross
- Department of Neurology, University Hospital Munster, Munster, Germany
- Department of Neurology, University of Munster, Munster, Germany
| | - Heinz Wiendl
- Department of Neurology, University Hospital Munster, Munster, Germany
- Department of Neurology, University of Munster, Munster, Germany
| | - Marieke Sm Van Ham
- Department of Immunopathology, Sanquin Research, Amsterdam, The Netherlands
- Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Anja Ten Brinke
- Department of Immunopathology, Sanquin Research, Amsterdam, The Netherlands
- Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Ana Maria Barriocanal
- Clinical Research Polyvalent Unit, Clinial Trial Unit-Spanish Clinical Research Network, Germans Trias i Pujol Health Sciences Research Institute, Badalona, Spain
- Department of Pharmacology, Therapeutic and Toxicology, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Anna Massuet-Vilamajó
- Institut de Diagnòstic per la Imatge, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Niel Hens
- Interuniversity Institute for Biostatistics and statistical Bioinformatics (I-BioStat), Universiteit Hasselt, Hasselt, Belgium
- Centre for Health Economic Research and Modelling Infectious Diseases, Vaccine & Infectious Disease Institute (VAXINFECTIO) & Center for Statistics, University of Antwerp Faculty of Medicine and Health Sciences, Wilrijk, Belgium
| | - Zwi Berneman
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp Faculty of Medicine and Health Sciences, Wilrijk, Belgium
- Center for Cell Therapy and Regenerative Medicine, University Hospital Antwerp, Edegem, Belgium
| | - Eva Martínez-Cáceres
- Division of Immunology, LCMN, Hospital Universitario Germans Trias i Pujol and Research Institute, Badalona, Spain
| | - Nathalie Cools
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp Faculty of Medicine and Health Sciences, Wilrijk, Belgium
- Center for Cell Therapy and Regenerative Medicine, University Hospital Antwerp, Edegem, Belgium
| | - Cristina Ramo-Tello
- Multiple Sclerosis Unit, Department of Neurosciences, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| |
Collapse
|
34
|
Derdelinckx J, Mansilla MJ, De Laere M, Lee WP, Navarro-Barriuso J, Wens I, Nkansah I, Daans J, De Reu H, Jolanta Keliris A, Van Audekerke J, Vanreusel V, Pieters Z, Van der Linden A, Verhoye M, Molenberghs G, Hens N, Goossens H, Willekens B, Cras P, Ponsaerts P, Berneman ZN, Martínez-Cáceres EM, Cools N. Clinical and immunological control of experimental autoimmune encephalomyelitis by tolerogenic dendritic cells loaded with MOG-encoding mRNA. J Neuroinflammation 2019; 16:167. [PMID: 31416452 PMCID: PMC6696692 DOI: 10.1186/s12974-019-1541-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 07/09/2019] [Indexed: 01/05/2023] Open
Abstract
Background Although effective in reducing relapse rate and delaying progression, current therapies for multiple sclerosis (MS) do not completely halt disease progression. T cell autoimmunity to myelin antigens is considered one of the main mechanisms driving MS. It is characterized by autoreactivity to disease-initiating myelin antigen epitope(s), followed by a cascade of epitope spreading, which are both strongly patient-dependent. Targeting a variety of MS-associated antigens by myelin antigen-presenting tolerogenic dendritic cells (tolDC) is a promising treatment strategy to re-establish tolerance in MS. Electroporation with mRNA encoding myelin proteins is an innovative technique to load tolDC with the full spectrum of naturally processed myelin-derived epitopes. Methods In this study, we generated murine tolDC presenting myelin oligodendrocyte glycoprotein (MOG) using mRNA electroporation and we assessed the efficacy of MOG mRNA-electroporated tolDC to dampen pathogenic T cell responses in experimental autoimmune encephalomyelitis (EAE). For this, MOG35–55-immunized C57BL/6 mice were injected intravenously at days 13, 17, and 21 post-disease induction with 1α,25-dihydroxyvitamin D3-treated tolDC electroporated with MOG-encoding mRNA. Mice were scored daily for signs of paralysis. At day 25, myelin reactivity was evaluated following restimulation of splenocytes with myelin-derived epitopes. Ex vivo magnetic resonance imaging (MRI) was performed to assess spinal cord inflammatory lesion load. Results Treatment of MOG35–55-immunized C57BL/6 mice with MOG mRNA-electroporated or MOG35–55-pulsed tolDC led to a stabilization of the EAE clinical score from the first administration onwards, whereas it worsened in mice treated with non-antigen-loaded tolDC or with vehicle only. In addition, MOG35–55-specific pro-inflammatory pathogenic T cell responses and myelin antigen epitope spreading were inhibited in the peripheral immune system of tolDC-treated mice. Finally, magnetic resonance imaging analysis of hyperintense spots along the spinal cord was in line with the clinical score. Conclusions Electroporation with mRNA is an efficient and versatile tool to generate myelin-presenting tolDC that are capable to stabilize the clinical score in EAE. These results pave the way for further research into mRNA-electroporated tolDC treatment as a patient-tailored therapy for MS. Electronic supplementary material The online version of this article (10.1186/s12974-019-1541-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Judith Derdelinckx
- Laboratory of Experimental Hematology, Faculty of Medicine and Health Sciences, Vaccine and Infectious Disease Institute (VaxInfectio), University of Antwerp, Antwerp University Hospital (UZA), Wilrijkstraat 10, 2650, Edegem, Belgium. .,Division of Neurology, Antwerp University Hospital, Edegem, Belgium.
| | - María José Mansilla
- Division of Immunology, Germans Trias i Pujol University Hospital and Research Institute, Campus Can Ruti, Badalona, Spain.,Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, Spain
| | - Maxime De Laere
- Laboratory of Experimental Hematology, Faculty of Medicine and Health Sciences, Vaccine and Infectious Disease Institute (VaxInfectio), University of Antwerp, Antwerp University Hospital (UZA), Wilrijkstraat 10, 2650, Edegem, Belgium
| | - Wai-Ping Lee
- Laboratory of Experimental Hematology, Faculty of Medicine and Health Sciences, Vaccine and Infectious Disease Institute (VaxInfectio), University of Antwerp, Antwerp University Hospital (UZA), Wilrijkstraat 10, 2650, Edegem, Belgium.,Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Juan Navarro-Barriuso
- Division of Immunology, Germans Trias i Pujol University Hospital and Research Institute, Campus Can Ruti, Badalona, Spain.,Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, Spain
| | - Inez Wens
- Laboratory of Experimental Hematology, Faculty of Medicine and Health Sciences, Vaccine and Infectious Disease Institute (VaxInfectio), University of Antwerp, Antwerp University Hospital (UZA), Wilrijkstraat 10, 2650, Edegem, Belgium
| | - Irene Nkansah
- Laboratory of Experimental Hematology, Faculty of Medicine and Health Sciences, Vaccine and Infectious Disease Institute (VaxInfectio), University of Antwerp, Antwerp University Hospital (UZA), Wilrijkstraat 10, 2650, Edegem, Belgium
| | - Jasmijn Daans
- Laboratory of Experimental Hematology, Faculty of Medicine and Health Sciences, Vaccine and Infectious Disease Institute (VaxInfectio), University of Antwerp, Antwerp University Hospital (UZA), Wilrijkstraat 10, 2650, Edegem, Belgium
| | - Hans De Reu
- Laboratory of Experimental Hematology, Faculty of Medicine and Health Sciences, Vaccine and Infectious Disease Institute (VaxInfectio), University of Antwerp, Antwerp University Hospital (UZA), Wilrijkstraat 10, 2650, Edegem, Belgium
| | | | | | | | - Zoë Pieters
- Center for Statistics, I-Biostat, Hasselt University, Diepenbeek, Belgium.,Centre for Health Economics Research and Modelling Infectious Diseases (CHERMID), Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | | | | | - Geert Molenberghs
- Center for Statistics, I-Biostat, Hasselt University, Diepenbeek, Belgium.,L-BioStat, I-BioStat, KU Leuven, Leuven, Belgium
| | - Niel Hens
- Center for Statistics, I-Biostat, Hasselt University, Diepenbeek, Belgium.,Centre for Health Economics Research and Modelling Infectious Diseases (CHERMID), Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Herman Goossens
- Laboratory of Medical Microbiology, Faculty of Medicine and Health Sciences, Vaccine and Infectious Disease Institute (VaxInfectio), University of Antwerp, Antwerp, Belgium
| | - Barbara Willekens
- Laboratory of Experimental Hematology, Faculty of Medicine and Health Sciences, Vaccine and Infectious Disease Institute (VaxInfectio), University of Antwerp, Antwerp University Hospital (UZA), Wilrijkstraat 10, 2650, Edegem, Belgium.,Division of Neurology, Antwerp University Hospital, Edegem, Belgium
| | - Patrick Cras
- Division of Neurology, Antwerp University Hospital, Edegem, Belgium.,Born Bunge Institute, Translational Neurosciences, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Peter Ponsaerts
- Laboratory of Experimental Hematology, Faculty of Medicine and Health Sciences, Vaccine and Infectious Disease Institute (VaxInfectio), University of Antwerp, Antwerp University Hospital (UZA), Wilrijkstraat 10, 2650, Edegem, Belgium
| | - Zwi N Berneman
- Laboratory of Experimental Hematology, Faculty of Medicine and Health Sciences, Vaccine and Infectious Disease Institute (VaxInfectio), University of Antwerp, Antwerp University Hospital (UZA), Wilrijkstraat 10, 2650, Edegem, Belgium.,Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Eva María Martínez-Cáceres
- Division of Immunology, Germans Trias i Pujol University Hospital and Research Institute, Campus Can Ruti, Badalona, Spain.,Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Cerdanyola del Vallès, Spain
| | - Nathalie Cools
- Laboratory of Experimental Hematology, Faculty of Medicine and Health Sciences, Vaccine and Infectious Disease Institute (VaxInfectio), University of Antwerp, Antwerp University Hospital (UZA), Wilrijkstraat 10, 2650, Edegem, Belgium.,Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium
| |
Collapse
|
35
|
Navarro-Barriuso J, Mansilla MJ, Quirant-Sánchez B, Ardiaca-Martínez A, Teniente-Serra A, Presas-Rodríguez S, ten Brinke A, Ramo-Tello C, Martínez-Cáceres EM. MAP7 and MUCL1 Are Biomarkers of Vitamin D3-Induced Tolerogenic Dendritic Cells in Multiple Sclerosis Patients. Front Immunol 2019; 10:1251. [PMID: 31293564 PMCID: PMC6598738 DOI: 10.3389/fimmu.2019.01251] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 05/16/2019] [Indexed: 12/14/2022] Open
Abstract
The administration of autologous tolerogenic dendritic cells (tolDC) has become a promising alternative for the treatment of autoimmune diseases, such as multiple sclerosis (MS). Specifically, the use of vitamin D3 for the generation of tolDC (vitD3-tolDC) constitutes one of the most widely studied approaches, as it has evidenced significant immune regulatory properties, both in vitro and in vivo. In this article, we generated human vitD3-tolDC from monocytes from healthy donors and MS patients, characterized in both cases by a semi-mature phenotype, secretion of IL-10 and inhibition of allogeneic lymphocyte proliferation. Additionally, we studied their transcriptomic profile and selected a number of differentially expressed genes compared to control mature and immature dendritic cells for their analysis. Among them, qPCR results validated CYP24A1, MAP7 and MUCL1 genes as biomarkers of vitD3-tolDC in both healthy donors and MS patients. Furthermore, we constructed a network of protein interactions based on the literature, which manifested that MAP7 and MUCL1 genes are both closely connected between them and involved in immune-related functions. In conclusion, this study evidences that MAP7 and MUCL1 constitute robust and potentially functional biomarkers of the generation of vitD3-tolDC, opening the window for their use as quality controls in clinical trials for MS.
Collapse
Affiliation(s)
- Juan Navarro-Barriuso
- Division of Immunology, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Barcelona, Spain
- Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - María José Mansilla
- Division of Immunology, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Barcelona, Spain
- Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Bibiana Quirant-Sánchez
- Division of Immunology, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Barcelona, Spain
- Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Alicia Ardiaca-Martínez
- Division of Immunology, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Barcelona, Spain
- Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Aina Teniente-Serra
- Division of Immunology, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Barcelona, Spain
- Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Silvia Presas-Rodríguez
- Multiple Sclerosis Unit, Department of Neurosciences, Germans Trias i Pujol University Hospital, Barcelona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Anja ten Brinke
- Department of Immunopathology, Sanquin Research, and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Cristina Ramo-Tello
- Multiple Sclerosis Unit, Department of Neurosciences, Germans Trias i Pujol University Hospital, Barcelona, Spain
| | - Eva M. Martínez-Cáceres
- Division of Immunology, LCMN, Germans Trias i Pujol University Hospital and Research Institute, Barcelona, Spain
- Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| |
Collapse
|
36
|
Immune tolerance in multiple sclerosis and neuromyelitis optica with peptide-loaded tolerogenic dendritic cells in a phase 1b trial. Proc Natl Acad Sci U S A 2019; 116:8463-8470. [PMID: 30962374 PMCID: PMC6486735 DOI: 10.1073/pnas.1820039116] [Citation(s) in RCA: 118] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Application of antigen-specific immune tolerance in autoimmune disease is a long-sought goal. We studied diseases with abundant information on the autoimmune target: in multiple sclerosis (MS), various myelin antigens are known targets of T cells and antibodies, whereas in neuromyelitis optica (NMO), the aquaporin-4 channel is attacked by T cells and antibodies. We tested whether engineered dendritic cells might induce a tolerogenic immune response in these two conditions. In this in-human clinical study, individual regulatory T cells, secreting IL-10, a key tolerogenic cytokine, were detected after treatment. These results might lead to more extensive trials with this approach in autoimmune conditions where the antigenic target has been identified, including MS, NMO, myasthenia gravis, and Graves disease. There are adaptive T-cell and antibody autoimmune responses to myelin-derived peptides in multiple sclerosis (MS) and to aquaporin-4 (AQP4) in neuromyelitis optica spectrum disorders (NMOSDs). Strategies aimed at antigen-specific tolerance to these autoantigens are thus indicated for these diseases. One approach involves induction of tolerance with engineered dendritic cells (tolDCs) loaded with specific antigens. We conducted an in-human phase 1b clinical trial testing increasing concentrations of autologous tolDCs loaded with peptides from various myelin proteins and from AQP4. We tested this approach in 12 patients, 8 with MS and 4 with NMOSD. The primary end point was the safety and tolerability, while secondary end points were clinical outcomes (relapses and disability), imaging (MRI and optical coherence tomography), and immunological responses. Therapy with tolDCs was well tolerated, without serious adverse events and with no therapy-related reactions. Patients remained stable clinically in terms of relapse, disability, and in various measurements using imaging. We observed a significant increase in the production of IL-10 levels in PBMCs stimulated with the peptides as well as an increase in the frequency of a regulatory T cell, known as Tr1, by week 12 of follow-up. In this phase 1b trial, we concluded that the i.v. administration of peptide-loaded dendritic cells is safe and feasible. Elicitation of specific IL-10 production by peptide-specific T cells in MS and NMOSD patients indicates that a key element in antigen specific tolerance is activated with this approach. The results warrant further clinical testing in larger trials.
Collapse
|
37
|
Beneficial effect of atorvastatin-modified dendritic cells pulsed with myelin oligodendrocyte glycoprotein autoantigen on experimental autoimmune encephalomyelitis. Neuroreport 2019; 29:317-327. [PMID: 29394220 DOI: 10.1097/wnr.0000000000000962] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
It is well known that dendritic cells play a key role in producing antigen-specific responses. Inversely, tolerogenic dendritic cells (TolDCs), a specialized subset, induce immune tolerance and negatively regulate autoimmune responses. Statins, the inhibitors of 3-hydroxy-3-methylglutaryl coenzyme A reductase in the mevalonate pathway for cholesterol biosynthesis, might be a promising inductive agent for inducing TolDCs. This study aimed to investigate the effectiveness of TolDCs induced by atorvastatin pulsed with myelin oligodendrocyte glycoprotein 35-55 peptide (MOG35-55) in experimental autoimmune encephalomyelitis mice established by MOG35-55 immunization and to investigate the potential effects on Th17/Treg balance in the murine model of multiple sclerosis. Our results showed that atorvastatin-treated dendritic cells maintained a steady semimature phenotype with a low level of costimulatory molecules and proinflammatory cytokines. Upon an intraperitoneal injection into experimental autoimmune encephalomyelitis mice, TolDCs pulsed with MOG (TolDCs-MOG) significantly alleviated disease activity and regulated Th17/Treg balance with a marked decrease in Th17 cells and an obvious increase in regulatory T cells. Taken together, TolDCs-MOG modified by atorvastatin showed a characteristic tolerogenic phenotype and the antigen-specific TolDCs might represent a new promising strategy for the future treatments for multiple sclerosis.
Collapse
|
38
|
Lewis JS, Stewart JM, Marshall GP, Carstens MR, Zhang Y, Dolgova NV, Xia C, Brusko TM, Wasserfall CH, Clare-Salzler MJ, Atkinson MA, Keselowsky BG. Dual-Sized Microparticle System for Generating Suppressive Dendritic Cells Prevents and Reverses Type 1 Diabetes in the Nonobese Diabetic Mouse Model. ACS Biomater Sci Eng 2019; 5:2631-2646. [PMID: 31119191 PMCID: PMC6518351 DOI: 10.1021/acsbiomaterials.9b00332] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 03/26/2019] [Indexed: 02/08/2023]
Abstract
![]()
Antigen
specificity is a primary goal in developing curative therapies
for autoimmune disease. Dendritic cells (DCs), as the most effective
antigen presenting cells in the body, represent a key target to mediate
restoration of antigen-specific immune regulation. Here, we describe
an injectable, dual-sized microparticle (MP) approach that employs
phagocytosable ∼1 μm and nonphagocytosable ∼30
μm MPs to deliver tolerance-promoting factors both intracellularly
and extracellularly, as well as the type 1 diabetes autoantigen, insulin,
to DCs for reprogramming of immune responses and remediation of autoimmunity.
This poly(lactic-co-glycolic acid) (PLGA) MP system
prevented diabetes onset in 60% of nonobese diabetic (NOD) mice when
administered subcutaneously in 8 week old mice. Prevention of disease
was dependent upon antigen inclusion and required encapsulation of
factors in MPs. Moreover, administration of this “suppressive-vaccine”
boosted pancreatic lymph node and splenic regulatory T cells (Tregs),
upregulated PD-1 on CD4+ and CD8+ T cells, and
reversed hyperglycemia for up to 100 days in recent-onset NOD mice.
Our results demonstrate that a MP-based platform can reeducate the
immune system in an antigen-specific manner, augment immunomodulation
compared to soluble administration of drugs, and provide a promising
alternative to systemic immunosuppression for autoimmunity.
Collapse
Affiliation(s)
- Jamal S Lewis
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 1275 Center Drive, Gainesville, Florida 32611, United States.,OneVax, LLC, 12085 Research Drive, Alachua, Florida 32615, United States.,Department of Biomedical Engineering, University of California-Davis, One Shields Avenue, Davis, California 95616, United States
| | - Joshua M Stewart
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 1275 Center Drive, Gainesville, Florida 32611, United States
| | - Gregory P Marshall
- OneVax, LLC, 12085 Research Drive, Alachua, Florida 32615, United States
| | - Matthew R Carstens
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 1275 Center Drive, Gainesville, Florida 32611, United States
| | - Ying Zhang
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 1275 Center Drive, Gainesville, Florida 32611, United States
| | - Natalia V Dolgova
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 1275 Center Drive, Gainesville, Florida 32611, United States
| | - Changqing Xia
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, 1600 SW Archer Road, Gainesville, Florida 32611, United States
| | - Todd M Brusko
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, 1600 SW Archer Road, Gainesville, Florida 32611, United States
| | - Clive H Wasserfall
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, 1600 SW Archer Road, Gainesville, Florida 32611, United States
| | - Michael J Clare-Salzler
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, 1600 SW Archer Road, Gainesville, Florida 32611, United States
| | - Mark A Atkinson
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, 1600 SW Archer Road, Gainesville, Florida 32611, United States.,Department of Pediatrics, University of Florida, 1600 SW Archer Road, Gainesville, Florida 32611, United States
| | - Benjamin G Keselowsky
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 1275 Center Drive, Gainesville, Florida 32611, United States.,Department of Pathology, Immunology and Laboratory Medicine, University of Florida, 1600 SW Archer Road, Gainesville, Florida 32611, United States
| |
Collapse
|
39
|
Ten Brinke A, Martinez-Llordella M, Cools N, Hilkens CMU, van Ham SM, Sawitzki B, Geissler EK, Lombardi G, Trzonkowski P, Martinez-Caceres E. Ways Forward for Tolerance-Inducing Cellular Therapies- an AFACTT Perspective. Front Immunol 2019; 10:181. [PMID: 30853957 PMCID: PMC6395407 DOI: 10.3389/fimmu.2019.00181] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 01/21/2019] [Indexed: 12/17/2022] Open
Abstract
Clinical studies with cellular therapies using tolerance-inducing cells, such as tolerogenic antigen-presenting cells (tolAPC) and regulatory T cells (Treg) for the prevention of transplant rejection and the treatment of autoimmune diseases have been expanding the last decade. In this perspective, we will summarize the current perspectives of the clinical application of both tolAPC and Treg, and will address future directions and the importance of immunomonitoring in clinical studies that will result in progress in the field.
Collapse
Affiliation(s)
- Anja Ten Brinke
- Department of Immunopathology, Sanquin Research, Amsterdam, Netherlands.,Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Marc Martinez-Llordella
- Department of Inflammation Biology, MRC Centre for Transplantation, School of Immunology and Microbial Sciences, Institute of Liver Studies, King's College London, London, United Kingdom
| | - Nathalie Cools
- Laboratory of Experimental Hematology, Faculty of Medicine and Health Sciences, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Catharien M U Hilkens
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - S Marieke van Ham
- Department of Immunopathology, Sanquin Research, Amsterdam, Netherlands.,Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands.,Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
| | - Birgit Sawitzki
- Charité-Universitaetsmedizin Berlin, Berlin Institute of Health, Institute for Medical Immunology, Humboldt-Universitaet zu Berlin, Berlin, Germany
| | - Edward K Geissler
- Section of Experimental Surgery, Department of Surgery, University Hospital Regensburg, University of Regensburg, Regensburg, Germany
| | - Giovanna Lombardi
- Division of Transplantation Immunology and Mucosal Biology, MRC Centre for Transplantation, Guy's Hospital, King's College London, London, United Kingdom
| | - Piotr Trzonkowski
- Department of Clinical Immunology and Transplantology, Medical University of Gdansk, Gdansk, Poland
| | - Eva Martinez-Caceres
- Division of Immunology, Germans Trias i Pujol University Hospital, LCMN, IGTP, Badalona, Spain.,Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| |
Collapse
|
40
|
Djedovic N, Jevtić B, Mansilla MJ, Petković F, Blaževski J, Timotijević G, Navarro-Barriuso J, Martinez-Caceres E, Mostarica Stojković M, Miljković Đ. Comparison of dendritic cells obtained from autoimmunty-prone and resistant rats. Immunobiology 2019; 224:470-476. [PMID: 30765133 DOI: 10.1016/j.imbio.2019.01.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 01/16/2019] [Indexed: 01/07/2023]
Abstract
Dendritic cells (DC) are responsible for the initiation and shaping of the adaptive immune response and are in the focus of autoimmunity research. We were interested in comparison of DC obtained from autoimmunity-prone Dark Agouti (DA) rats and autoimmunity-resistant Albino Oxford (AO) rats. DC were generated from bone marrow precursors and matured (mDC) by lipopolysaccharide. Tolerogenic DC (tolDC) obtained by vitamin D3 treatment were studied in parallel. Profile of cytokine production was different in AO and DA mDC and tolDC. Expression of MHC class II molecules and CD86 were higher in DA DC, while vitamin D3 reduced their expression in dendritic cells of both strains. Allogeneic proliferation of CD4+ T cells was reduced by AO tolDC, but not with DA tolDC in comparison to respective mDC. Finally, expression of various genes identified as differentially expressed in human mDC and tolDC was also analyzed in AO and DA DC. Again, AO and DA DC differed in the expression of the analyzed genes. To conclude, AO and DA DC differ in production of cytokines, expression of antigen presentation-related molecules and in regulation of CD4+ T proliferation. The difference is valuable for understanding the divergence of the strains in their susceptibility to autoimmunity.
Collapse
Affiliation(s)
- Neda Djedovic
- Department of Immunology, Institute for Biological Research "Siniša Stanković", University of Belgrade, Serbia
| | - Bojan Jevtić
- Department of Immunology, Institute for Biological Research "Siniša Stanković", University of Belgrade, Serbia
| | - M José Mansilla
- Immunology Division, Germans Trias i Pujol University Hospital and Research Institute, Badalona, Spain; Department of Cellular Biology, Physiology, and Immunology, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Filip Petković
- Department of Immunology, Institute for Biological Research "Siniša Stanković", University of Belgrade, Serbia
| | - Jana Blaževski
- Department of Immunology, Institute for Biological Research "Siniša Stanković", University of Belgrade, Serbia
| | - Gordana Timotijević
- Laboratory for Plant Molecular Biology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Serbia
| | - Juan Navarro-Barriuso
- Immunology Division, Germans Trias i Pujol University Hospital and Research Institute, Badalona, Spain; Department of Cellular Biology, Physiology, and Immunology, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Spain
| | - Eva Martinez-Caceres
- Immunology Division, Germans Trias i Pujol University Hospital and Research Institute, Badalona, Spain
| | | | - Đorđe Miljković
- Department of Immunology, Institute for Biological Research "Siniša Stanković", University of Belgrade, Serbia.
| |
Collapse
|
41
|
DE Wolf C, VAN DE Bovenkamp M, Hoefnagel M. Regulatory perspective on in vitro potency assays for human dendritic cells used in anti-tumor immunotherapy. Cytotherapy 2018; 20:1289-1308. [PMID: 30327247 DOI: 10.1016/j.jcyt.2018.07.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 05/25/2018] [Accepted: 07/14/2018] [Indexed: 12/18/2022]
Abstract
Dendritic cells (DCs) are key connectors between the innate and adaptive immune system and have an important role in modulating other immune cells. Therefore, their therapeutic application to steer immune responses is considered in various disorders, including cancer. Due to differences in the cell source and manufacturing process, each DC medicinal product is unique. Consequently, release tests to ensure consistent quality need to be product-specific. Although general guidance concerning quality control testing of cell-based therapies is available, cell type-specific regulation is still limited. Especially guidance related to potency testing is needed, because developing an in vitro assay measuring cell properties relevant for in vivo functionality is challenging. In this review, we provide DC-specific guidance for development of in vitro potency assays for characterisation and release. We present a broad overview of in vitro potency assays suggested for DC products to determine their anti-tumor functionality. Several advantages and limitations of these assays are discussed. Also, we provide some points to consider for selection and design of a potency test. The ideal functionality assay for anti-tumor products evaluates the capacity of DCs to stimulate antigen-specific T cells. Because this approach may not be feasible for release, use of surrogate potency markers could be considered, provided that these markers are sufficiently linked to the in vivo DC biological activity and clinical response. Further elucidation of the involvement of specific DC subsets in anti-tumor responses will result in improved manufacturing processes for DC-based products and should be considered during potency assay development.
Collapse
Affiliation(s)
- Charlotte DE Wolf
- Medicines Evaluation Board College ter Beoordeling van Geneesmiddelen-Medicines Evaluation Board (CBG-MEB), Utrecht, The Netherlands; Department of Infectious Diseases and Immunology, Utrecht University, The Netherlands
| | - Marja VAN DE Bovenkamp
- Medicines Evaluation Board College ter Beoordeling van Geneesmiddelen-Medicines Evaluation Board (CBG-MEB), Utrecht, The Netherlands
| | - Marcel Hoefnagel
- Medicines Evaluation Board College ter Beoordeling van Geneesmiddelen-Medicines Evaluation Board (CBG-MEB), Utrecht, The Netherlands.
| |
Collapse
|
42
|
Navarro-Barriuso J, Mansilla MJ, Martínez-Cáceres EM. Searching for the Transcriptomic Signature of Immune Tolerance Induction-Biomarkers of Safety and Functionality for Tolerogenic Dendritic Cells and Regulatory Macrophages. Front Immunol 2018; 9:2062. [PMID: 30298066 PMCID: PMC6160751 DOI: 10.3389/fimmu.2018.02062] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 08/21/2018] [Indexed: 12/12/2022] Open
Abstract
The last years have witnessed a breakthrough in the development of cell-based tolerance-inducing cell therapies for the treatment of autoimmune diseases and solid-organ transplantation. Indeed, the use of tolerogenic dendritic cells (tolDC) and regulatory macrophages (Mreg) is currently being tested in Phase I and Phase II clinical trials worldwide, with the aim of finding an effective therapy able to abrogate the inflammatory processes causing these pathologies without compromising the protective immunity of the patients. However, there exists a wide variety of different protocols to generate human tolDC and Mreg and, consequently, the characteristics of each product are heterogeneous. For this reason, the identification of biomarkers able to define their functionality (tolerogenicity) is of great relevance, on the one hand, to guarantee the safety of tolDC and Mreg before administration and, on the other hand, to compare the results between different cell products and laboratories. In this article, we perform an exhaustive review of protocols generating human tolDC and Mreg in the literature, aiming to elucidate if there are any common transcriptomic signature or potential biomarkers of tolerogenicity among the different approaches. However, and although several effectors seem to be induced in common in some of the most reported protocols to generate both tolDC or Mreg, the transcriptomic profile of these cellular products strongly varies depending on the approach used to generate them.
Collapse
Affiliation(s)
- Juan Navarro-Barriuso
- Division of Immunology, Germans Trias i Pujol University Hospital and Research Institute, Barcelona, Spain.,Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - María José Mansilla
- Division of Immunology, Germans Trias i Pujol University Hospital and Research Institute, Barcelona, Spain.,Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Eva M Martínez-Cáceres
- Division of Immunology, Germans Trias i Pujol University Hospital and Research Institute, Barcelona, Spain.,Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
43
|
Pei W, Wan X, Shahzad KA, Zhang L, Song S, Jin X, Wang L, Zhao C, Shen C. Direct modulation of myelin-autoreactive CD4 + and CD8 + T cells in EAE mice by a tolerogenic nanoparticle co-carrying myelin peptide-loaded major histocompatibility complexes, CD47 and multiple regulatory molecules. Int J Nanomedicine 2018; 13:3731-3750. [PMID: 29983566 PMCID: PMC6027825 DOI: 10.2147/ijn.s164500] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Purpose Numerous nanomaterials have been reported in the treatment of multiple sclerosis or experimental autoimmune encephalomyelitis (EAE). But most of these nanoscale therapeutics deliver myelin antigens together with toxins or cytokines and underlay the cellular uptake and induction of tolerogenic antigen-presenting cells by which they indirectly induce T cell tolerance. This study focuses on the on-target and direct modulation of myelin-autoreactive T cells and combined use of multiple regulatory molecules by generating a tolerogenic nanoparticle. Materials and methods Poly(lactic-co-glycolic acid) nanoparticles (PLGA-NPs) were fabricated by co-coupling MOG40–54/H-2Db-Ig dimer, MOG35–55/I-Ab multimer, anti-Fas, PD-L1-Fc and CD47-Fc and encapsulating transforming growth factor-β1. The resulting 217 nm tolerogenic nanoparticles (tNPs) were administered intravenously into MOG35–55 peptide-induced EAE mice, which was followed by the investigation of therapeutic outcomes and the in vivo mechanism. Results Four infusions of the tNPs durably ameliorated EAE with a marked reduction of clinical score, neuroinflammation and demyelination. They were distributed in secondary lymphoid tissues, various organs and brain after intravenous injection, with retention over 36 h, and made contacts with CD4+ and CD8+ T cells. Two injections of the tNPs markedly decreased the MOG35–55-reactive Th1 and Th17 cells and MOG40–55-reactive Tc1 and Tc17 cells, increased regulatory T cells, inhibited T cell proliferation and elevated T cell apoptosis in spleen. Transforming growth factor-β1 and interleukin-10 were upregulated in the homogenates of central nervous system and supernatant of spleen cells. Conclusion Our data suggest a novel therapeutic nanoparticle to directly modulate autoreactive T cells by surface presentation of multiple ligands and paracrine release of cytokine in the antigen-specific combination immunotherapy for T cell-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Weiya Pei
- Department of Microbiology and Immunology, Medical School, Southeast University, Nanjing, Jiangsu 210009, China,
| | - Xin Wan
- Department of Microbiology and Immunology, Medical School, Southeast University, Nanjing, Jiangsu 210009, China,
| | - Khawar Ali Shahzad
- Department of Microbiology and Immunology, Medical School, Southeast University, Nanjing, Jiangsu 210009, China,
| | - Lei Zhang
- Department of Microbiology and Immunology, Medical School, Southeast University, Nanjing, Jiangsu 210009, China,
| | - Shilong Song
- Department of Microbiology and Immunology, Medical School, Southeast University, Nanjing, Jiangsu 210009, China,
| | - Xiaoxiao Jin
- Department of Microbiology and Immunology, Medical School, Southeast University, Nanjing, Jiangsu 210009, China,
| | - Limin Wang
- Department of Microbiology and Immunology, Medical School, Southeast University, Nanjing, Jiangsu 210009, China,
| | - Chen Zhao
- Department of Microbiology and Immunology, Medical School, Southeast University, Nanjing, Jiangsu 210009, China,
| | - Chuanlai Shen
- Department of Microbiology and Immunology, Medical School, Southeast University, Nanjing, Jiangsu 210009, China,
| |
Collapse
|
44
|
Flórez-Grau G, Zubizarreta I, Cabezón R, Villoslada P, Benitez-Ribas D. Tolerogenic Dendritic Cells as a Promising Antigen-Specific Therapy in the Treatment of Multiple Sclerosis and Neuromyelitis Optica From Preclinical to Clinical Trials. Front Immunol 2018; 9:1169. [PMID: 29904379 PMCID: PMC5990597 DOI: 10.3389/fimmu.2018.01169] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 05/09/2018] [Indexed: 12/20/2022] Open
Abstract
The identification of activated T-lymphocytes restricted to myelin-derived immunogenic peptides in multiple sclerosis (MS) and aquaporin-4 water channel in neuromyelitis optica (NMO) in the blood of patients opened the possibility for developing highly selective and disease-specific therapeutic approaches. Antigen presenting cells and in particular dendritic cells (DCs) represent a strategy to inhibit pro-inflammatory T helper cells. DCs are located in peripheral and lymphoid tissues and are essential for homeostasis of T cell-dependent immune responses. The expression of a particular set of receptors involved in pathogen recognition confers to DCs the property to initiate immune responses. However, in the absence of danger signals different DC subsets have been revealed to induce active tolerance by inducing regulatory T cells, inhibiting pro-inflammatory T helper cells responses or both. Interestingly, several protocols to generate clinical-grade tolerogenic DC (Tol-DC) in vitro have been described, offering the possibility to restore the homeostasis to central nervous system-related antigens. In this review, we discuss about different DC subsets and their role in tolerance induction, the different protocols to generate Tol-DCs and preclinical studies in animal models as well as describe recent characterization of Tol-DCs for clinical application in autoimmune diseases and in particular in MS and NMO patients. In addition, we discuss the clinical trials ongoing based on Tol-DCs to treat different autoimmune diseases.
Collapse
Affiliation(s)
- Georgina Flórez-Grau
- Department of Immunology, Hospital Clinic i Provincial, Barcelona, Spain.,Neuroimmunology Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Irati Zubizarreta
- Neuroimmunology Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Raquel Cabezón
- Department of Immunology, Hospital Clinic i Provincial, Barcelona, Spain.,Neuroimmunology Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Pablo Villoslada
- Neuroimmunology Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | | |
Collapse
|
45
|
Abstract
Over the past century, solid organ transplantation has been improved both at a surgical and postoperative level. However, despite the improvement in efficiency, safety, and survival, we are still far from obtaining full acceptance of all kinds of allograft in the absence of concomitant treatments. Today, transplanted patients are treated with immunosuppressive drugs (IS) to minimize immunological response in order to prevent graft rejection. Nevertheless, the lack of specificity of IS leads to an increase in the risk of cancer and infections. At this point, cell therapies have been shown as a novel promising resource to minimize the use of IS in transplantation. The main strength of cell therapy is the opportunity to generate allograft-specific tolerance, promoting in this way long-term allograft survival. Among several other regulatory cell types, tolerogenic monocyte-derived dendritic cells (Tol-MoDCs) appear to be an interesting candidate for cell therapy due to their ability to perform specific antigen presentation and to polarize immune response to immunotolerance. In this review, we describe the characteristics and the mechanisms of action of both human Tol-MoDCs and rodent tolerogenic bone marrow-derived DCs (Tol-BMDCs). Furthermore, studies performed in transplantation models in rodents and non-human primates corroborate the potential of Tol-BMDCs for immunoregulation. In consequence, Tol-MoDCs have been recently evaluated in sundry clinical trials in autoimmune diseases and shown to be safe. In addition to autoimmune diseases clinical trials, Tol-MoDC is currently used in the first phase I/II clinical trials in transplantation. Translation of Tol-MoDCs to clinical application in transplantation will also be discussed in this review.
Collapse
Affiliation(s)
- Eros Marín
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Nephrologie (ITUN), CHU Nantes, Nantes, France
| | - Maria Cristina Cuturi
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Nephrologie (ITUN), CHU Nantes, Nantes, France
| | - Aurélie Moreau
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Nephrologie (ITUN), CHU Nantes, Nantes, France
| |
Collapse
|
46
|
Funda DP, Goliáš J, Hudcovic T, Kozáková H, Špíšek R, Palová-Jelínková L. Antigen Loading (e.g., Glutamic Acid Decarboxylase 65) of Tolerogenic DCs (tolDCs) Reduces Their Capacity to Prevent Diabetes in the Non-Obese Diabetes (NOD)-Severe Combined Immunodeficiency Model of Adoptive Cotransfer of Diabetes As Well As in NOD Mice. Front Immunol 2018; 9:290. [PMID: 29503651 PMCID: PMC5820308 DOI: 10.3389/fimmu.2018.00290] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 02/01/2018] [Indexed: 12/12/2022] Open
Abstract
Tolerogenic DCs (tolDCs) are being researched as a promising intervention strategy also in autoimmune diseases including type 1 diabetes (T1D). T1D is a T-cell-mediated, organ-specific disease with several well-defined and rather specific autoantigens, i.e., proinsulin, insulin, glutamic acid decarboxylase 65 (GAD65), that have been used in animal as well as human intervention trials in attempts to achieve a more efficient, specific immunotherapy. In this study, we have tested tolerogenic DCs for their effectiveness to prevent adoptive transfer of diabetes by diabetogenic splenocytes into non-obese diabetes (NOD)-severe combined immunodeficiency (NOD-SCID) recipients. While i.p. application of tolDCs prepared from bone marrow of prediabetic NOD mice by vitamin D2 and dexamethasone significantly reduced diabetes transfer into the NOD-SCID females, this effect was completely abolished when tolDCs were loaded with the mouse recombinant GAD65, but also with a control protein—ovalbumin (OVA). The effect was not dependent on the presence of serum in the tolDC culture. Similar results were observed in NOD mice. Removal of possible bystander antigen-presenting cells within the diabetogenic splenocytes by negative magnetic sorting of T cells did not alter this surprising effect. Tolerogenic DCs loaded with an immunodominant mouse GAD65 peptide also displayed diminished diabetes-preventive effect. Tolerogenic DCs were characterized by surface maturation markers (CD40, CD80, CD86, MHC II) and the lipopolysaccharide stability test. Data from alloreactive T cell proliferation and cytokine induction assays (IFN-γ) did not reveal the differences observed in the diabetes incidence. Migration of tolDCs, tolDCs-GAD65 and tolDCs-OVA to spleen, mesenteric- and pancreatic lymph nodes displayed similar, mucosal pattern with highest accumulation in pancreatic lymph nodes present up to 9 days after the i.p. application. These data document that mechanisms by which tolDCs operate in vivo require much better understanding for improving efficacy of this promising cell therapy, especially in the presence of an antigen, e.g., GAD65.
Collapse
Affiliation(s)
- David P Funda
- Institute of Microbiology of the Czech Academy of Sciences, v.v.i., Prague, Czechia
| | - Jaroslav Goliáš
- Institute of Microbiology of the Czech Academy of Sciences, v.v.i., Prague, Czechia
| | - Tomáš Hudcovic
- Institute of Microbiology of the Czech Academy of Sciences, v.v.i., Nový Hrádek, Czechia
| | - Hana Kozáková
- Institute of Microbiology of the Czech Academy of Sciences, v.v.i., Nový Hrádek, Czechia
| | - Radek Špíšek
- SOTIO a s., Prague, Czechia.,Department of Immunology, 2nd Medical School, Charles University, Prague, Czechia
| | - Lenka Palová-Jelínková
- SOTIO a s., Prague, Czechia.,Department of Immunology, 2nd Medical School, Charles University, Prague, Czechia
| |
Collapse
|
47
|
De Laere M, Derdelinckx J, Hassi M, Kerosalo M, Oravamäki H, Van den Bergh J, Berneman Z, Cools N. Shuttling Tolerogenic Dendritic Cells across the Blood-Brain Barrier In Vitro via the Introduction of De Novo C-C Chemokine Receptor 5 Expression Using Messenger RNA Electroporation. Front Immunol 2018; 8:1964. [PMID: 29403473 PMCID: PMC5778265 DOI: 10.3389/fimmu.2017.01964] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 12/19/2017] [Indexed: 01/06/2023] Open
Abstract
The use of tolerance-inducing dendritic cells (tolDCs) has been proven to be safe and well tolerated in the treatment of autoimmune diseases. Nevertheless, several challenges remain, including finding ways to facilitate the migration of cell therapeutic products to lymph nodes, and the site of inflammation. In the treatment of neuroinflammatory diseases, such as multiple sclerosis (MS), the blood-brain barrier (BBB) represents a major obstacle to the delivery of therapeutic agents to the inflamed central nervous system (CNS). As it was previously demonstrated that C-C chemokine receptor 5 (CCR5) may be involved in inflammatory migration of DCs, the aim of this study was to investigate CCR5-driven migration of tolDCs. Only a minority of in vitro generated vitamin D3 (vitD3)-treated tolDCs expressed the inflammatory chemokine receptor CCR5. Thus, messenger RNA (mRNA) encoding CCR5 was introduced by means of electroporation (EP). After mRNA EP, tolDCs transiently displayed increased levels of CCR5 protein expression. Accordingly, the capacity of mRNA electroporated tolDCs to transmigrate toward a chemokine gradient in an in vitro model of the BBB improved significantly. Neither the tolerogenic phenotype nor the T cell-stimulatory function of tolDCs was affected by mRNA EP. EP of tolDCs with mRNA encoding CCR5 enabled these cells to migrate to inflammatory sites. The approach used herein has important implications for the treatment of MS. Using this approach, tolDCs actively shuttle across the BBB, allowing in situ down-modulation of autoimmune responses in the CNS.
Collapse
Affiliation(s)
- Maxime De Laere
- Laboratory of Experimental Hematology, Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Wilrijk, Belgium
| | - Judith Derdelinckx
- Laboratory of Experimental Hematology, Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Wilrijk, Belgium.,Department of Neurology, Antwerp University Hospital, Edegem, Belgium
| | - Mari Hassi
- Laboratory of Experimental Hematology, Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Wilrijk, Belgium
| | - Mari Kerosalo
- Laboratory of Experimental Hematology, Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Wilrijk, Belgium
| | - Heidi Oravamäki
- Laboratory of Experimental Hematology, Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Wilrijk, Belgium
| | - Johan Van den Bergh
- Laboratory of Experimental Hematology, Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Wilrijk, Belgium
| | - Zwi Berneman
- Laboratory of Experimental Hematology, Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Wilrijk, Belgium.,Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Nathalie Cools
- Laboratory of Experimental Hematology, Faculty of Medicine and Health Sciences, Vaccine & Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Wilrijk, Belgium
| |
Collapse
|
48
|
Abstract
Multiple sclerosis (MS) is a chronic immune-mediated disease of the central nervous system (CNS) characterized by neuroinflammation, neurodegeneration and impaired repair mechanisms that lead to neurological disability. The crux of MS is the patient's own immune cells attacking self-antigens in the CNS, namely the myelin sheath that protects nerve cells of the brain and spinal cord. Restoring antigen-specific tolerance via therapeutic vaccination is an innovative and exciting approach in MS therapy. Indeed, leveraging the body's attempt to prevent autoimmunity, i.e., tolerization, focuses on the underlying cause of the disease and could be the key to solving neuroinflammation. In this perspective, antigen-specific vaccination targets only the detrimental and aberrant immune response against the specific disease-associated antigen(s) involved while retaining the capacity of the immune system to respond to unrelated antigens. We review the experimental approaches of tolerance-inducing vaccination in relapsing and progressive forms of MS that have reached the clinical development phase, including vaccination with autologous T cells, autologous tolerogenic dendritic cells, T cell receptor peptide vaccination, altered peptide ligand, ATX-MS-1467, cluster of differentiation (CD)-206-targeted liposomal myelin basic protein peptides and DNA vaccination. Failures, successes and future directions are discussed.
Collapse
|
49
|
van Eden W, Jansen MAA, de Wolf ACM, Ludwig IS, Leufkens P, Broere F. The Immunomodulatory Potential of tolDCs Loaded with Heat Shock Proteins. Front Immunol 2017; 8:1690. [PMID: 29250070 PMCID: PMC5717764 DOI: 10.3389/fimmu.2017.01690] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 11/16/2017] [Indexed: 01/24/2023] Open
Abstract
Disease suppressive T cell regulation may depend on cognate interactions of regulatory T cells with self-antigens that are abundantly expressed in the inflamed tissues. Heat shock proteins (HSPs) are by their nature upregulated in stressed cells and therefore abundantly present as potential targets for such regulation. HSP immunizations have led to inhibition of experimentally induced inflammatory conditions in various models. However, re-establishment of tolerance in the presence of an ongoing inflammatory process has remained challenging. Since tolerogenic DCs (tolDCs) have the combined capacity of mitigating antigen-specific inflammatory responses and of endowing T cells with regulatory potential, it seems attractive to combine the anti-inflammatory qualities of tolDCs with those of HSPs.
Collapse
Affiliation(s)
- Willem van Eden
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Manon A A Jansen
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - A Charlotte Mt de Wolf
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Irene S Ludwig
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | | | - Femke Broere
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
50
|
Pujol-Autonell I, Mansilla MJ, Rodriguez-Fernandez S, Cano-Sarabia M, Navarro-Barriuso J, Ampudia RM, Rius A, Garcia-Jimeno S, Perna-Barrull D, Martinez-Caceres E, Maspoch D, Vives-Pi M. Liposome-based immunotherapy against autoimmune diseases: therapeutic effect on multiple sclerosis. Nanomedicine (Lond) 2017; 12:1231-1242. [PMID: 28593827 DOI: 10.2217/nnm-2016-0410] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM Based on the ability of apoptosis to induce immunological tolerance, liposomes were generated mimicking apoptotic cells, and they arrest autoimmunity in Type 1 diabetes. Our aim was to validate the immunotherapy in other autoimmune disease: multiple sclerosis. MATERIALS & METHODS Phosphatidylserine-rich liposomes were loaded with disease-specific autoantigen. Therapeutic capability of liposomes was assessed in vitro and in vivo. RESULTS Liposomes induced a tolerogenic phenotype in dendritic cells, and arrested autoimmunity, thus decreasing the incidence, delaying the onset and reducing the severity of experimental disease, correlating with an increase in a probably regulatory CD25+ FoxP3- CD4+ T-cell subset. CONCLUSION This is the first work that confirms phosphatidylserine-liposomes as a powerful tool to arrest multiple sclerosis, demonstrating its relevance for clinical application.
Collapse
Affiliation(s)
- Irma Pujol-Autonell
- Immunology Division, Germans Trias i Pujol University Hospital & Research Institute, Department of Cellular Biology, Physiology & Immunology, Autonomous University of Barcelona, 08916 Badalona, Spain
| | - Maria-Jose Mansilla
- Immunology Division, Germans Trias i Pujol University Hospital & Research Institute, Department of Cellular Biology, Physiology & Immunology, Autonomous University of Barcelona, 08916 Badalona, Spain
| | - Silvia Rodriguez-Fernandez
- Immunology Division, Germans Trias i Pujol University Hospital & Research Institute, Department of Cellular Biology, Physiology & Immunology, Autonomous University of Barcelona, 08916 Badalona, Spain
| | - Mary Cano-Sarabia
- Catalan Institute of Nanoscience & Nanotechnology (ICN2), CSIC & The Barcelona Institute of Science & Technology, 08193 Bellaterra, Barcelona, Spain
| | - Juan Navarro-Barriuso
- Immunology Division, Germans Trias i Pujol University Hospital & Research Institute, Department of Cellular Biology, Physiology & Immunology, Autonomous University of Barcelona, 08916 Badalona, Spain
| | - Rosa-Maria Ampudia
- Immunology Division, Germans Trias i Pujol University Hospital & Research Institute, Department of Cellular Biology, Physiology & Immunology, Autonomous University of Barcelona, 08916 Badalona, Spain
| | - Aleix Rius
- Immunology Division, Germans Trias i Pujol University Hospital & Research Institute, Department of Cellular Biology, Physiology & Immunology, Autonomous University of Barcelona, 08916 Badalona, Spain
| | - Sonia Garcia-Jimeno
- Catalan Institute of Nanoscience & Nanotechnology (ICN2), CSIC & The Barcelona Institute of Science & Technology, 08193 Bellaterra, Barcelona, Spain
| | - David Perna-Barrull
- Immunology Division, Germans Trias i Pujol University Hospital & Research Institute, Department of Cellular Biology, Physiology & Immunology, Autonomous University of Barcelona, 08916 Badalona, Spain
| | - Eva Martinez-Caceres
- Immunology Division, Germans Trias i Pujol University Hospital & Research Institute, Department of Cellular Biology, Physiology & Immunology, Autonomous University of Barcelona, 08916 Badalona, Spain
| | - Daniel Maspoch
- Catalan Institute of Nanoscience & Nanotechnology (ICN2), CSIC & The Barcelona Institute of Science & Technology, 08193 Bellaterra, Barcelona, Spain.,Institucio Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
| | - Marta Vives-Pi
- Immunology Division, Germans Trias i Pujol University Hospital & Research Institute, Department of Cellular Biology, Physiology & Immunology, Autonomous University of Barcelona, 08916 Badalona, Spain.,CIBER of Diabetes & Associated Metabolic Diseases (CIBERDEM), ISCIII, Madrid, Spain
| |
Collapse
|