1
|
Doelman W, Reijnen RC, Dijksman N, Janssen APA, van Driel N, 't Hart BA, Philippens I, Araman C, Baron W, van Kasteren SI. Citrullinated human and murine MOG 35-55 display distinct biophysical and biochemical behavior. J Biol Chem 2023; 299:103065. [PMID: 36841486 PMCID: PMC10060747 DOI: 10.1016/j.jbc.2023.103065] [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: 01/08/2023] [Revised: 02/15/2023] [Accepted: 02/18/2023] [Indexed: 02/27/2023] Open
Abstract
The peptide spanning residues 35 to 55 of the protein myelin oligodendrocyte glycoprotein (MOG) has been studied extensively in its role as a key autoantigen in the neuroinflammatory autoimmune disease multiple sclerosis. Rodents and nonhuman primate species immunized with this peptide develop a neuroinflammatory condition called experimental autoimmune encephalomyelitis, often used as a model for multiple sclerosis. Over the last decade, the role of citrullination of this antigen in the disease onset and progression has come under increased scrutiny. We recently reported on the ability of these citrullinated MOG35-55 peptides to aggregate in an amyloid-like fashion, suggesting a new potential pathogenic mechanism underlying this disease. The immunodominant region of MOG is highly conserved between species, with the only difference between the murine and human protein, a polymorphism on position 42, which is serine in mice and proline for humans. Here, we show that the biophysical and biochemical behavior we previously observed for citrullinated murine MOG35-55 is fundamentally different for human and mouse MOG35-55. The citrullinated human peptides do not show amyloid-like behavior under the conditions where the murine peptides do. Moreover, we tested the ability of these peptides to stimulate lymphocytes derived from MOG immunized marmoset monkeys. While the citrullinated murine peptides did not produce a proliferative response, one of the citrullinated human peptides did. We postulate that this unexpected difference is caused by disparate antigen processing. Taken together, our results suggest that further study on the role of citrullination in MOG-induced experimental autoimmune encephalomyelitis is necessary.
Collapse
Affiliation(s)
- W Doelman
- Department of Bio-Organic Synthesis, Leiden Institute of Chemistry, Leiden, the Netherlands
| | - R C Reijnen
- Department of Bio-Organic Synthesis, Leiden Institute of Chemistry, Leiden, the Netherlands
| | - N Dijksman
- Section Molecular Neurobiology, Department Biomedical Sciences of Cells & Systems, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - A P A Janssen
- Department of Bio-Organic Synthesis, Leiden Institute of Chemistry, Leiden, the Netherlands
| | - N van Driel
- Department of Immunobiology, Biomedical Primate Research Center, Rijswijk, the Netherlands
| | - B A 't Hart
- Section Molecular Neurobiology, Department Biomedical Sciences of Cells & Systems, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - I Philippens
- Department of Immunobiology, Biomedical Primate Research Center, Rijswijk, the Netherlands
| | - C Araman
- Department of Bio-Organic Synthesis, Leiden Institute of Chemistry, Leiden, the Netherlands
| | - W Baron
- Section Molecular Neurobiology, Department Biomedical Sciences of Cells & Systems, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands.
| | - S I van Kasteren
- Department of Bio-Organic Synthesis, Leiden Institute of Chemistry, Leiden, the Netherlands.
| |
Collapse
|
2
|
Lopez JA, Denkova M, Ramanathan S, Dale RC, Brilot F. Pathogenesis of autoimmune demyelination: from multiple sclerosis to neuromyelitis optica spectrum disorders and myelin oligodendrocyte glycoprotein antibody-associated disease. Clin Transl Immunology 2021; 10:e1316. [PMID: 34336206 PMCID: PMC8312887 DOI: 10.1002/cti2.1316] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/20/2021] [Accepted: 07/01/2021] [Indexed: 12/16/2022] Open
Abstract
Autoimmunity plays a significant role in the pathogenesis of demyelination. Multiple sclerosis (MS), neuromyelitis optica spectrum disorders (NMOSD) and myelin oligodendrocyte glycoprotein antibody‐associated disease (MOGAD) are now recognised as separate disease entities under the amalgam of human central nervous system demyelinating disorders. While these disorders share inherent similarities, investigations into their distinct clinical presentations and lesion pathologies have aided in differential diagnoses and understanding of disease pathogenesis. An interplay of various genetic and environmental factors contributes to each disease, many of which implicate an autoimmune response. The pivotal role of the adaptive immune system has been highlighted by the diagnostic autoantibodies in NMOSD and MOGAD, and the presence of autoreactive lymphocytes in MS lesions. While a number of autoantigens have been proposed in MS, recent emphasis on the contribution of B cells has shed new light on the well‐established understanding of T cell involvement in pathogenesis. This review aims to synthesise the clinical characteristics and pathological findings, discuss existing and emerging hypotheses regarding the aetiology of demyelination and evaluate recent pathogenicity studies involving T cells, B cells, and autoantibodies and their implications in human demyelination.
Collapse
Affiliation(s)
- Joseph A Lopez
- Brain Autoimmunity Group Kids Neuroscience Centre Kids Research at the Children's Hospital at Westmead Sydney NSW Australia.,Specialty of Child and Adolescent Health Faculty of Medicine and Health The University of Sydney Sydney NSW Australia
| | - Martina Denkova
- Brain Autoimmunity Group Kids Neuroscience Centre Kids Research at the Children's Hospital at Westmead Sydney NSW Australia.,School of Medical Sciences Faculty of Medicine and Health The University of Sydney Sydney NSW Australia
| | - Sudarshini Ramanathan
- Brain Autoimmunity Group Kids Neuroscience Centre Kids Research at the Children's Hospital at Westmead Sydney NSW Australia.,Sydney Medical School Faculty of Medicine and Health The University of Sydney Sydney NSW Australia.,Department of Neurology Concord Hospital Sydney NSW Australia
| | - Russell C Dale
- Brain Autoimmunity Group Kids Neuroscience Centre Kids Research at the Children's Hospital at Westmead Sydney NSW Australia.,Specialty of Child and Adolescent Health Faculty of Medicine and Health The University of Sydney Sydney NSW Australia.,Sydney Medical School Faculty of Medicine and Health The University of Sydney Sydney NSW Australia.,Brain and Mind Centre The University of Sydney Sydney NSW Australia
| | - Fabienne Brilot
- Brain Autoimmunity Group Kids Neuroscience Centre Kids Research at the Children's Hospital at Westmead Sydney NSW Australia.,Specialty of Child and Adolescent Health Faculty of Medicine and Health The University of Sydney Sydney NSW Australia.,School of Medical Sciences Faculty of Medicine and Health The University of Sydney Sydney NSW Australia.,Brain and Mind Centre The University of Sydney Sydney NSW Australia
| |
Collapse
|
3
|
Cruz-Herranz A, Dietrich M, Hilla AM, Yiu HH, Levin MH, Hecker C, Issberner A, Hallenberger A, Cordano C, Lehmann-Horn K, Balk LJ, Aktas O, Ingwersen J, von Gall C, Hartung HP, Zamvil SS, Fischer D, Albrecht P, Green AJ. Monitoring retinal changes with optical coherence tomography predicts neuronal loss in experimental autoimmune encephalomyelitis. J Neuroinflammation 2019; 16:203. [PMID: 31684959 PMCID: PMC6827223 DOI: 10.1186/s12974-019-1583-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 09/10/2019] [Indexed: 02/06/2023] Open
Abstract
Background Retinal optical coherence tomography (OCT) is a clinical and research tool in multiple sclerosis, where it has shown significant retinal nerve fiber (RNFL) and ganglion cell (RGC) layer thinning, while postmortem studies have reported RGC loss. Although retinal pathology in experimental autoimmune encephalomyelitis (EAE) has been described, comparative OCT studies among EAE models are scarce. Furthermore, the best practices for the implementation of OCT in the EAE lab, especially with afoveate animals like rodents, remain undefined. We aimed to describe the dynamics of retinal injury in different mouse EAE models and outline the optimal experimental conditions, scan protocols, and analysis methods, comparing these to histology to confirm the pathological underpinnings. Methods Using spectral-domain OCT, we analyzed the test-retest and the inter-rater reliability of volume, peripapillary, and combined horizontal and vertical line scans. We then monitored the thickness of the retinal layers in different EAE models: in wild-type (WT) C57Bl/6J mice immunized with myelin oligodendrocyte glycoprotein peptide (MOG35–55) or with bovine myelin basic protein (MBP), in TCR2D2 mice immunized with MOG35–55, and in SJL/J mice immunized with myelin proteolipid lipoprotein (PLP139–151). Strain-matched control mice were sham-immunized. RGC density was counted on retinal flatmounts at the end of each experiment. Results Volume scans centered on the optic disc showed the best reliability. Retinal changes during EAE were localized in the inner retinal layers (IRLs, the combination of the RNFL and the ganglion cell plus the inner plexiform layers). In WT, MOG35–55 EAE, progressive thinning of IRL started rapidly after EAE onset, with 1/3 of total loss occurring during the initial 2 months. IRL thinning was associated with the degree of RGC loss and the severity of EAE. Sham-immunized SJL/J mice showed progressive IRL atrophy, which was accentuated in PLP-immunized mice. MOG35–55-immunized TCR2D2 mice showed severe EAE and retinal thinning. MBP immunization led to very mild disease without significant retinopathy. Conclusions Retinal neuroaxonal damage develops quickly during EAE. Changes in retinal thickness mirror neuronal loss and clinical severity. Monitoring of the IRL thickness after immunization against MOG35–55 in C57Bl/6J mice seems the most convenient model to study retinal neurodegeneration in EAE.
Collapse
Affiliation(s)
- Andrés Cruz-Herranz
- Division of Neuroimmunology and Glial Biology, Department of Neurology, University of California, San Francisco, San Francisco, USA
| | - Michael Dietrich
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Alexander M Hilla
- Department of Cell Physiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Bochum, Germany
| | - Hao H Yiu
- Division of Neuroimmunology and Glial Biology, Department of Neurology, University of California, San Francisco, San Francisco, USA
| | - Marc H Levin
- Department of Ophthalmology, University of California, San Francisco, San Francisco, USA.,Department of Ophthalmology, Palo Alto Medical Foundation, Palo Alto, CA, USA
| | - Christina Hecker
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Andrea Issberner
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Angelika Hallenberger
- Institute of Anatomy II, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Christian Cordano
- Division of Neuroimmunology and Glial Biology, Department of Neurology, University of California, San Francisco, San Francisco, USA
| | - Klaus Lehmann-Horn
- Department of Neurology, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Lisanne J Balk
- Department of Neurology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Orhan Aktas
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Jens Ingwersen
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Charlotte von Gall
- Institute of Anatomy II, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Scott S Zamvil
- Division of Neuroimmunology and Glial Biology, Department of Neurology, University of California, San Francisco, San Francisco, USA.,Program in Immunology, University of California, San Francisco, San Francisco, USA
| | - Dietmar Fischer
- Department of Cell Physiology, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Bochum, Germany
| | - Philipp Albrecht
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany.
| | - Ari J Green
- Division of Neuroimmunology and Glial Biology, Department of Neurology, University of California, San Francisco, San Francisco, USA. .,Department of Ophthalmology, University of California, San Francisco, San Francisco, USA.
| |
Collapse
|
4
|
Hori J, Yamaguchi T, Keino H, Hamrah P, Maruyama K. Immune privilege in corneal transplantation. Prog Retin Eye Res 2019; 72:100758. [PMID: 31014973 DOI: 10.1016/j.preteyeres.2019.04.002] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 04/14/2019] [Accepted: 04/16/2019] [Indexed: 12/13/2022]
Abstract
Corneal transplantation is the most successful solid organ transplantation performed in humans. The extraordinary success of orthotopic corneal allografts, in both humans and experimental animals, is related to the phenomenon of "immune privilege". Inflammation is self-regulated to preserve ocular functions because the eye has immune privilege. At present, three major mechanisms are considered to provide immune privilege in corneal transplantation: 1) anatomical, cellular, and molecular barriers in the cornea; 2) tolerance related to anterior chamber-associated immune deviation and regulatory T cells; and 3) an immunosuppressive intraocular microenvironment. This review describes the mechanisms of immune privilege that have been elucidated from animal models of ocular inflammation, especially those involving corneal transplantation, and its relevance for the clinic. An update on molecular, cellular, and neural interactions in local and systemic immune regulation is provided. Therapeutic strategies for restoring immune privilege are also discussed.
Collapse
Affiliation(s)
- Junko Hori
- Department of Ophthalmology, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo, 113-8603, Japan; Department of Ophthalmology, Nippon Medical School, Tama-Nagayama Hospital, 1-7-1 Nagayama, Tama, Tokyo, 206-8512, Japan.
| | - Takefumi Yamaguchi
- Department of Ophthalmology, Tokyo Dental College Ichikawa General Hospital, 5-11-13 Sugano, Ichikawa-shi, Chiba, 272-8513, Japan; Department of Ophthalmology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Hiroshi Keino
- Department of Ophthalmology, Kyorin University School of Medicine, 6-20-2 Shinkawa, Mitaka-shi, Tokyo, 181-8611, Japan
| | - Pedram Hamrah
- Center for Translational Ocular Immunology, Tufts Medical Center, Tufts University School of Medicine, Tufts University, 800 Washington St, Boston, MA, 02111, USA; Department of Ophthalmology, Tufts Medical Center, Tufts University School of Medicine, Tufts University, 800 Washington St, Boston, MA, 02111, USA
| | - Kazuichi Maruyama
- Department of Innovative Visual Science, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
5
|
Shah NM, Lai PF, Imami N, Johnson MR. Progesterone-Related Immune Modulation of Pregnancy and Labor. Front Endocrinol (Lausanne) 2019; 10:198. [PMID: 30984115 PMCID: PMC6449726 DOI: 10.3389/fendo.2019.00198] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 03/11/2019] [Indexed: 12/17/2022] Open
Abstract
Pregnancy involves a complex interplay between maternal neuroendocrine and immunological systems in order to establish and sustain a growing fetus. It is thought that the uterus at pregnancy transitions from quiescent to laboring state in response to interactions between maternal and fetal systems at least partly via altered neuroendocrine signaling. Progesterone (P4) is a vital hormone in maternal reproductive tissues and immune cells during pregnancy. As such, P4 is widely used in clinical interventions to improve the chance of embryo implantation, as well as reduce the risk of miscarriage and premature labor. Here we review research to date that focus on the pathways through which P4 mediates its actions on both the maternal reproductive and immune system. We will dissect the role of P4 as a modulator of inflammation, both systemic and intrinsic to the uterus, during human pregnancy and labor.
Collapse
Affiliation(s)
- Nishel M. Shah
- Department of Surgery and Cancer, Chelsea and Westminster Hospital, Imperial College London, London, United Kingdom
| | - Pei F. Lai
- Department of Surgery and Cancer, Chelsea and Westminster Hospital, Imperial College London, London, United Kingdom
| | - Nesrina Imami
- Department of Medicine, Chelsea and Westminster Hospital, Imperial College London, London, United Kingdom
| | - Mark R. Johnson
- Department of Surgery and Cancer, Chelsea and Westminster Hospital, Imperial College London, London, United Kingdom
| |
Collapse
|
6
|
MohanKrishnan A, Patel H, Bhurani V, Parmar R, Yadav N, Dave N, Rana S, Gupta S, Madariya J, Vyas P, Dalai SK. Inclusion of non-target antigen in vaccination favors generation of OVA specific CD4 memory T cells. Cell Immunol 2019; 337:1-14. [PMID: 30773218 DOI: 10.1016/j.cellimm.2018.11.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Revised: 10/04/2018] [Accepted: 11/19/2018] [Indexed: 11/19/2022]
Abstract
Inducing long-lived memory T cells by sub-unit vaccines has been a challenge. Subunit vaccines containing single immunogenic target antigen from a given pathogen have been designed with the presumption of mimicking the condition associated with natural infection, but fail to induce quality memory responses. In this study, we have included non-target antigens with vaccine candidate, OVA, in the inoculum containing TLR ligands to suffice the minimal condition of pathogen to provoke immune response. We found that inclusion of immunogenic HEL (hen egg lysozyme) or poorly immunogenic MBP (Myelin Basic protein) non-target antigen enhances the OVA specific CD4 T cell responses. Interestingly, poorly immunogenic MBP was found to strongly favor the generation of OVA specific memory CD4 T cells. MBP not only improves magnitude of T cell response but also promotes the T cells to undergo higher cycles of division, one of the characteristic of central memory T cells. Inclusion of MBP with vaccine targets was also found to promote multiple cytokine producing CD4 T cells. We also found that challenge of host with non-target antigen MBP favors generation of central Memory T cells.
Collapse
Affiliation(s)
| | - Hardik Patel
- Institute of Science, Nirma University, Ahmedabad, Gujarat, India
| | - Vishakha Bhurani
- Institute of Science, Nirma University, Ahmedabad, Gujarat, India
| | - Rajesh Parmar
- Institute of Science, Nirma University, Ahmedabad, Gujarat, India
| | - Naveen Yadav
- Institute of Science, Nirma University, Ahmedabad, Gujarat, India
| | - Niyam Dave
- Institute of Science, Nirma University, Ahmedabad, Gujarat, India
| | - Sonal Rana
- Institute of Science, Nirma University, Ahmedabad, Gujarat, India
| | - Somnath Gupta
- Institute of Science, Nirma University, Ahmedabad, Gujarat, India
| | - Jagdish Madariya
- Institute of Science, Nirma University, Ahmedabad, Gujarat, India
| | - Prerak Vyas
- Institute of Science, Nirma University, Ahmedabad, Gujarat, India
| | | |
Collapse
|
7
|
Alvites-Misajel K, García-Gutiérrez M, Miranda-Rodríguez C, Ramos-Escudero F. Organically vs conventionally-grown dark and white chia seeds ( Salvia hispanica L.): fatty acid composition, antioxidant activity and techno-functional properties. GRASAS Y ACEITES 2019. [DOI: 10.3989/gya.0462181] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The effects of organic and conventional crop systems on chemical composition, antioxidant activity and functional properties were evaluated in white and dark chia (Salvia hispanica L.) seeds. The organic system reduced the total protein content, and increased the total carbohydrates but did not change polyunsaturated fatty acids, total phenolic or flavonoids. Organic white chia seeds showed the best techno-functional properties. The antioxidant capacity of chia extracts varied in relation to the chemical complexity and differential rate kinetics of different assays. Extractable total phenolic acids and antioxidant capacity were better in organic white chia seeds. In this first approach, we have demonstrated that the organic white chia seed has a better total antioxidant capacity measured by direct quencher approaches than its conventionally-grown counterpart. To summarize, we conclude that the organic white chia seed could be a dietary source of antioxidants with a potential to promote health benefits in systemic functions and/or microbiota and the use of its techno-functional properties for the food industry.
Collapse
|
8
|
Švajger U, Rožman P. Induction of Tolerogenic Dendritic Cells by Endogenous Biomolecules: An Update. Front Immunol 2018; 9:2482. [PMID: 30416505 PMCID: PMC6212600 DOI: 10.3389/fimmu.2018.02482] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 10/08/2018] [Indexed: 12/19/2022] Open
Abstract
The importance of microenvironment on dendritic cell (DC) function and development has been strongly established during the last two decades. Although DCs with general tolerogenic characteristics have been isolated and defined as a particular sub-population, it is predominantly their unequivocal biological plasticity, which allows for unparalleled responsiveness to environmental ques and shaping of their tolerogenic characteristics when interacting with tolerance-inducing biomolecules. Dendritic cells carry receptors for a great number of endogenous factors, which, after ligation, can importantly influence the development of their activation state. For this there is ample evidence merely by observation of DC characteristics isolated from various anatomical niches, e.g., the greater immunosuppressive potential of DCs isolated from intestine compared to conventional blood DCs. Endogenous biomolecules present in these environments most likely play a major role as a determinant of their phenotype and function. In this review, we will concisely summarize in what way various, tolerance-inducing endogenous factors influence DC biology, the development of their particular tolerogenic state and their subsequent actions in context of immune response inhibition and induction of regulatory T cells.
Collapse
Affiliation(s)
- Urban Švajger
- Department for Therapeutic Services, Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia.,Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Primož Rožman
- Department for Therapeutic Services, Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia
| |
Collapse
|
9
|
Ahmed MS, Bae YS. Dendritic Cell-based Immunotherapy for Rheumatoid Arthritis: from Bench to Bedside. Immune Netw 2016; 16:44-51. [PMID: 26937231 PMCID: PMC4770099 DOI: 10.4110/in.2016.16.1.44] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 01/30/2016] [Accepted: 02/02/2016] [Indexed: 02/07/2023] Open
Abstract
Dendritic cells (DCs) are professional antigen presenting cells, and play an important role in the induction of antigen-specific adaptive immunity. However, some DC populations are involved in immune regulation and immune tolerance. These DC populations are believed to take part in the control of immune exaggeration and immune disorder, and maintain immune homeostasis in the body. Tolerogenic DCs (tolDCs) can be generated in vitro by genetic or pharmacological modification or by controlling the maturation stages of cytokine-derived DCs. These tolDCs have been investigated for the treatment of rheumatoid arthritis (RA) in experimental animal models. In the last decade, several in vitro and in vivo approaches have been translated into clinical trials. As of 2015, three tolDC trials for RA are on the list of ClinicalTrial.gov (www.clinicaltrials.gov). Other trials for RA are in progress and will be listed soon. In this review, we discuss the evolution of tolDC-based immunotherapy for RA and its limitations and future prospects.
Collapse
Affiliation(s)
- Md Selim Ahmed
- Department of Biological Science, Sungkyunkwan University, Suwon 16419, Korea
| | - Yong-Soo Bae
- Department of Biological Science, Sungkyunkwan University, Suwon 16419, Korea
| |
Collapse
|
10
|
Yuan R, Wang B, Lu W, Maeda Y, Dowling P. A Distinct Region in Erythropoietin that Induces Immuno/Inflammatory Modulation and Tissue Protection. Neurotherapeutics 2015; 12:850-61. [PMID: 26271954 PMCID: PMC4604189 DOI: 10.1007/s13311-015-0379-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Beneficial effects of short-term whole-molecule erythropoietin (EPO) therapy have been demonstrated on several animal models of diverse central nervous system pathology. However, the increased hematocrit induced by EPO-driven marrow stimulation greatly limits its potential for side effect-free therapy. We created a library of EPO-derived fragments based on the hypothesis that 2 distinct functions, erythropoiesis and tissue protection, reside in different regions of the molecule. Several small EPO-derived peptides within the Aβ loop of whole EPO molecule were screened for tissue protection in EAE mice. The 19-mer JM-4 peptide that contains 2 cysteine molecules consistently demonstrated the most potent clinical beneficial effects without producing hematocrit alterations in animal models of EAE. The JM-4-induced tissue protection was associated with modulation of the immunoregulatory process that drives inflammation and provokes subsequent autoimmune damage. Like the whole EPO molecule, JM-4 effectively modulated immune/inflammatory reaction within both the peripheral lymphatic tissue and central nervous system. The major effects induced by JM-4 include blocked expansion of monocyte/dendritic antigen presenting cell and T helper 17 cell populations, decreased proinflammatory cytokine production, and sharply enhanced expansion of the regulatory T-cell population. JM-4 shows promise for treatment of a broad spectrum of neural and non-neural conditions associated with inflammation.
Collapse
Affiliation(s)
- RuiRong Yuan
- Neurology Service, VA Medical Center of East Orange, East Orange, NJ, USA
- Department of Neurology and Neurosciences, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Bo Wang
- Neurology Service, VA Medical Center of East Orange, East Orange, NJ, USA
- Department of Neurology and Neurosciences, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Wei Lu
- Neurology Service, VA Medical Center of East Orange, East Orange, NJ, USA
| | - Yasuhiro Maeda
- Neurology Service, VA Medical Center of East Orange, East Orange, NJ, USA
- Department of Neurology and Neurosciences, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Peter Dowling
- Neurology Service, VA Medical Center of East Orange, East Orange, NJ, USA.
- Department of Neurology and Neurosciences, Rutgers New Jersey Medical School, Newark, NJ, USA.
| |
Collapse
|
11
|
Glenn JD, Smith MD, Kirby LA, Baxi EG, Whartenby KA. Disparate Effects of Mesenchymal Stem Cells in Experimental Autoimmune Encephalomyelitis and Cuprizone-Induced Demyelination. PLoS One 2015; 10:e0139008. [PMID: 26407166 PMCID: PMC4583481 DOI: 10.1371/journal.pone.0139008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 09/07/2015] [Indexed: 01/10/2023] Open
Abstract
Mesenchymal stem cells (MSCs) are pleiotropic cells with potential therapeutic benefits for a wide range of diseases. Because of their immunomodulatory properties they have been utilized to treat autoimmune diseases such as multiple sclerosis (MS), which is characterized by demyelination. The microenvironment surrounding MSCs is thought to affect their differentiation and phenotype, which could in turn affect the efficacy. We thus sought to dissect the potential for differential impact of MSCs on central nervous system (CNS) disease in T cell mediated and non-T cell mediated settings using the MOG35–55 experimental autoimmune encephalomyelitis (EAE) and cuprizone-mediated demyelination models, respectively. As the pathogeneses of MS and EAE are thought to be mediated by IFNγ-producing (TH1) and IL-17A-producing (TH17) effector CD4+ T cells, we investigated the effect of MSCs on the development of these two key pathogenic cell groups. Although MSCs suppressed the activation and effector function of TH17 cells, they did not affect TH1 activation, but enhanced TH1 effector function and ultimately produced no effect on EAE. In the non- T cell mediated cuprizone model of demyelination, MSC administration had a positive effect, with an overall increase in myelin abundance in the brain of MSC-treated mice compared to controls. These results highlight the potential variability of MSCs as a biologic therapeutic tool in the treatment of autoimmune disease and the need for further investigation into the multifaceted functions of MSCs in diverse microenvironments and the mechanisms behind the diversity.
Collapse
Affiliation(s)
- Justin D. Glenn
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Matthew D. Smith
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Leslie A. Kirby
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Emily G. Baxi
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Katharine A Whartenby
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
12
|
Duraes FV, Lippens C, Steinbach K, Dubrot J, Brighouse D, Bendriss-Vermare N, Issazadeh-Navikas S, Merkler D, Hugues S. pDC therapy induces recovery from EAE by recruiting endogenous pDC to sites of CNS inflammation. J Autoimmun 2015; 67:8-18. [PMID: 26341385 PMCID: PMC4758828 DOI: 10.1016/j.jaut.2015.08.014] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 08/19/2015] [Accepted: 08/20/2015] [Indexed: 11/25/2022]
Abstract
Plasmacytoid dendritic cells (pDCs) exhibit both innate and adaptive functions. In particular they are the main source of type I IFNs and directly impact T cell responses through antigen presentation. We have previously demonstrated that during experimental autoimmune encephalomyelitis (EAE) initiation, myelin-antigen presentation by pDCs is associated with suppressive Treg development and results in attenuated EAE. Here, we show that pDCs transferred during acute disease phase confer recovery from EAE. Clinical improvement is associated with migration of injected pDCs into inflamed CNS and is dependent on the subsequent and selective chemerin-mediated recruitment of endogenous pDCs to the CNS. The protective effect requires pDC pre-loading with myelin antigen, and is associated with the modulation of CNS-infiltrating pDC phenotype and inhibition of CNS encephalitogenic T cells. This study may pave the way for novel pDC-based cell therapies in autoimmune diseases, aiming at specifically modulating pathogenic cells that induce and sustain autoimmune inflammation. pDC therapy ameliorates established EAE. CNS inflammation is locally modulated after pDC transfer. Upon pDC transfer, resting endogenous pDCs are selectively recruited to the CNS via chemerin/CMKLR1 axis. Therapeutic pDC injection promotes a tolerogenic environment and inhibits encephalitogenic T cells in the CNS.
Collapse
Affiliation(s)
- Fernanda V Duraes
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva 4, Switzerland
| | - Carla Lippens
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva 4, Switzerland
| | - Karin Steinbach
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva 4, Switzerland
| | - Juan Dubrot
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva 4, Switzerland
| | - Dale Brighouse
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva 4, Switzerland
| | - Nathalie Bendriss-Vermare
- Université Lyon 1, INSERM U1052, CNRS, UMR5286, Centre de Recherche en Cancérologie de Lyon, Centre Léon Bérard, LabEx DEVweCAN, Lyon, France
| | | | - Doron Merkler
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva 4, Switzerland; Department of Pathology and Immunology, Division of Clinical Pathology, University & University Hospital of Geneva, Switzerland
| | - Stephanie Hugues
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva 4, Switzerland.
| |
Collapse
|
13
|
Wu HJ, Lo Y, Luk D, Lau CS, Lu L, Mok MY. Alternatively activated dendritic cells derived from systemic lupus erythematosus patients have tolerogenic phenotype and function. Clin Immunol 2014; 156:43-57. [PMID: 25463431 DOI: 10.1016/j.clim.2014.10.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2014] [Revised: 10/10/2014] [Accepted: 10/28/2014] [Indexed: 12/20/2022]
Abstract
Tolerogenic dendritic cells (DCs) are potential cell-based therapy in autoimmune diseases. In this study, we generated alternatively activated DCs (aaDCs) by treating monocyte-derived DCs from patients with systemic lupus erythematosus (SLE) and healthy subjects with combination of 1,25 dihydroxyvitamin D(3) (vitD3) and dexamethasone followed by lipopolysaccharide-induced maturation. Lupus aaDCs were found to acquire semi-mature phenotype that remained maturation-resistant to immunostimulants. They produced low level of IL-12 but high level of IL-10. They had attenuated allostimulatory effects on T cell activation and proliferation comparable to normal aaDCs and demonstrated differential immunomodulatory effects on naïve and memory T cells. These aaDCs were capable of inducing IL-10 producing regulatory T effectors from naïve T cells whereas they modulated cytokine profile with suppressed production of IFN-γ and IL-17 by co-cultured memory T cells with attenuated proliferation. These aaDCs were shown to be superior to those generated using vitD3 alone in lupus patients.
Collapse
Affiliation(s)
- Hai Jing Wu
- Division of Rheumatology & Clinical Immunology, Department of Medicine, University of Hong Kong, Hong Kong.
| | - Yi Lo
- Division of Rheumatology & Clinical Immunology, Department of Medicine, University of Hong Kong, Hong Kong.
| | - Daniel Luk
- Division of Rheumatology & Clinical Immunology, Department of Medicine, University of Hong Kong, Hong Kong.
| | - Chak Sing Lau
- Division of Rheumatology & Clinical Immunology, Department of Medicine, University of Hong Kong, Hong Kong.
| | - Liwei Lu
- Department of Pathology, University of Hong Kong, Hong Kong.
| | - Mo Yin Mok
- Division of Rheumatology & Clinical Immunology, Department of Medicine, University of Hong Kong, Hong Kong.
| |
Collapse
|
14
|
Hsu SM, Mathew R, Taylor AW, Stein-Streilein J. Ex-vivo tolerogenic F4/80⁺ antigen-presenting cells (APC) induce efferent CD8⁺ regulatory T cell-dependent suppression of experimental autoimmune uveitis. Clin Exp Immunol 2014; 176:37-48. [PMID: 24266626 PMCID: PMC3958152 DOI: 10.1111/cei.12243] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/19/2013] [Indexed: 12/23/2022] Open
Abstract
It is known that inoculation of antigen into the anterior chamber (a.c.) of a mouse eye induces a.c.-associated immune deviation (ACAID), which is mediated in part by antigen-specific local and peripheral tolerance to the inciting antigen. ACAID can also be induced in vivo by intravenous (i.v.) inoculation of ex-vivo-generated tolerogenic antigen-presenting cells (TolAPC). The purpose of this study was to test if in-vitro-generated retinal antigen-pulsed TolAPC suppressed established experimental autoimmune uveitis (EAU). Retinal antigen-pulsed TolAPC were injected i.v. into mice 7 days post-induction of EAU. We observed that retinal antigen-pulsed TolAPC suppressed the incidence and severity of the clinical expression of EAU and reduced the expression of associated inflammatory cytokines. Moreover, extract of whole retina efficiently replaced interphotoreceptor retinoid-binding protein (IRBP) in the preparation of TolAPC used to induce tolerance in EAU mice. Finally, the suppression of EAU could be transferred to a new set of EAU mice with CD8+ but not with CD4+regulatory T cells (Treg). Retinal antigen-pulsed TolAPC suppressed ongoing EAU by inducing CD8+ Treg cells that, in turn, suppressed the effector activity of the IRBP-specific T cells and altered the clinical symptoms of autoimmune inflammation in the eye. The ability to use retinal extract for the antigen raises the possibility that retinal extract could be used to produce autologous TolAPC and then used as therapy in human uveitis.
Collapse
Affiliation(s)
- S-M Hsu
- Schepens Eye Research Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; Department of Ophthalmology, National Cheng-Kung University Hospital, Tainan City, Taiwan
| | | | | | | |
Collapse
|
15
|
Zheng D, Wen L, Li C, Peng A, Cao Q, Wang Y, Harris D. Adoptive transfer of bone marrow dendritic cells failed to localize in the renal cortex and to improve renal injury in adriamycin nephropathy. Nephron Clin Pract 2014; 126:8-15. [PMID: 24526139 DOI: 10.1159/000358086] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Accepted: 12/12/2013] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND AND AIMS Murine bone marrow (BM) dendritic cells (DCs) can be modulated to be tolerogenic by cytokines, such as interleukin (IL)-10 and transforming growth factor (TGF)-β, and may play a regulatory role and sustain immune hemostasis in cognate kidney disease. However, it is unknown whether BM-DCs can be used to protect against renal injury in murine Adriamycin nephropathy (AN). METHODS In this study, by adoptive in vivo transfer of BM-DCs, including immature DCs, mature DCs (lipopolysaccharide-stimulated DCs) and BM regulatory DCs (IL-10/TGF-β-modified DCs, DCregs), we addressed the potential benefits of BM-DCs in chronic kidney disease. RESULTS We found that after adoptive transfer of DCregs, renal injury, including glomerulosclerosis, interstitial fibrosis and tubular atrophy, was not changed compared to AN controls. Correspondingly, renal functions measured by serum creatinine, 12-hour urine protein and creatinine clearance were also not improved by transfusion with DCregs compared to AN controls. CONCLUSION This study showed that the adoptive transfer of BM-DCs was unable to improve renal injury in an AN model, and this failure related to their inability to access the kidney.
Collapse
Affiliation(s)
- Dong Zheng
- Department of Nephrology, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai, China
| | | | | | | | | | | | | |
Collapse
|
16
|
Willett K, Bennett J. Immunology of AAV-Mediated Gene Transfer in the Eye. Front Immunol 2013; 4:261. [PMID: 24009613 PMCID: PMC3757345 DOI: 10.3389/fimmu.2013.00261] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2013] [Accepted: 08/16/2013] [Indexed: 12/20/2022] Open
Abstract
The eye has been at the forefront of translational gene therapy largely owing to suitable disease targets, anatomic accessibility, and well-studied immunologic privilege. These advantages have fostered research culminating in several clinical trials and adeno-associated virus (AAV) has emerged as the vector of choice for many ocular therapies. Pre-clinical and clinical investigations have assessed the humoral and cellular immune responses to a variety of naturally occurring and engineered AAV serotypes as well as their delivered transgenes and these data have been correlated to potential clinical sequelae. Encouragingly, AAV appears safe and effective with clinical follow-up surpassing 5 years in some studies. As disease targets continue to expand for AAV in the eye, thorough and deliberate assessment of immunologic safety is critical. With careful study, the development of these technologies should concurrently inform the biology of the ocular immune response.
Collapse
Affiliation(s)
- Keirnan Willett
- Department of Ophthalmology, Scheie Eye Institute, F.M. Kirby Center for Molecular Ophthalmology, University of Pennsylvania , Philadelphia, PA , USA
| | | |
Collapse
|
17
|
Abstract
INTRODUCTION CD8(+) T cells were originally considered to exert a suppressive role in demyelinating disease because of bias toward the CD4(+) T cell-mediated experimental autoimmune encephalomyelitis, the most common multiple sclerosis (MS) model. However, recent studies of human MS lesion samples and cerebrospinal fluid (CSF) provided compelling evidence about the pathogenic role of CD8(+) T cells. In this article, we discuss the theoretical roles of different CD8(+) T-cell subsets in MS. AREAS COVERED A revised focus from CD4(+) to CD8(+) T cell-mediated demyelinating disease is summarized. Clonal expansion of CD8(+) T cells in MS lesions and in vitro evidence that CD8(+) T cells injure every central nervous system (CNS) cell type and transect axons are discussed. The role of CD8(+) T cells in two animal models of MS and of regulatory, interleukin (IL)-17-secreting CD8(+) T cells is reviewed. Lastly, an overview about the pathogenic and/or beneficial role of various CD8(+) T-cell subsets is offered. EXPERT OPINION Growing evidence supports the pathogenic role of CD8(+) T cells. Clonally expanded CD8(+) T cells within MS lesions may damage the nervous system. Revealing the specific antigen is critical to design novel efficient treatments with minimal adverse effects. Increasing evidence exists for the role of regulatory, IL-17-secreting CD8(+) T cells in MS.
Collapse
|
18
|
Farooq SM, Ashour HM. Eye-mediated induction of specific immune tolerance to encephalitogenic antigens. CNS Neurosci Ther 2013; 19:503-10. [PMID: 23522052 DOI: 10.1111/cns.12087] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Revised: 02/13/2013] [Accepted: 02/16/2013] [Indexed: 01/27/2023] Open
Abstract
AIMS Administration of antigens into the anterior chamber (AC) of the eye induces a form of antigen-specific immune tolerance termed anterior chamber-associated immune deviation (ACAID). This immune tolerance effectively impairs host delayed-type hypersensitivity (DTH) responses. We hypothesized that ACAID could be generated in BALB/c mice following AC inoculation of the encephalitogenic antigens myelin oligodendrocyte glycoprotein (MOG) and myelin basic protein (MBP). METHODS We used DTH assays and local adoptive transfer (LAT) assays to test whether MOG/MBP-induced ACAID following their administration into the AC, whether they elicited this immune tolerance via CD8(+) T cells, and whether their AC coadministration (MOG/MBP) induced specific immune tolerance to one or both antigens. RESULTS We showed that MOG/MBP-induced AC-mediated specific immune tolerance, as evident from impaired DTH responses. This antigen-driven DTH suppression was solely mediated via splenic CD8(+) T cells as confirmed by LAT assays. Finally, a single AC injection with both antigens was sufficient to induce specific immune tolerance to these antigens, as evident from DTH and LAT assays. CONCLUSION ACAID T-cell regulation could be used as a therapeutic tool in the treatment of complicated autoimmune diseases that involve multiple antigens such as multiple sclerosis.
Collapse
Affiliation(s)
- Shukkur M Farooq
- Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | | |
Collapse
|
19
|
Type II collagen induces peripheral tolerance in BALB/c mice via the generation of CD8+ T regulatory cells. PLoS One 2012; 7:e48635. [PMID: 23133648 PMCID: PMC3487721 DOI: 10.1371/journal.pone.0048635] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Accepted: 10/03/2012] [Indexed: 11/19/2022] Open
Abstract
Antigens introduced into the anterior chamber (AC) of the eye induce a potent form of antigen-specific peripheral immune tolerance termed AC-associated immune deviation (ACAID), which prevents inflammatory immune responses and is characterized by impaired delayed-type hypersensitivity (DTH) responses. Type-II collagen (CII) is a fibrillar protein expressed exclusively in cartilage tissues. Although of its clinical relevance to Rheumatoid arthritis, aging, and osteoarthritis, there have been no studies to date to test if CII has the ability to induce ACAID. We hypothesized that ACAID could be generated via AC injection of CII in BALB/c mice. Using a DTH assay, the hypothesis was supported and led to another hypothesis that CII is capable of inducing specific immune tolerance via CD8(+) T regulatory cells (Tregs). Thus, we performed functional local adoptive transfer (LAT) assays to examine the regulatory roles of spleen cells, T cells, and CD8(+) T cells in the specific immune regulation induced by CII injection into the AC. Results indicated that CII induced ACAID when injected into the AC. Spleen cells of mice injected with CII in the AC significantly suppressed DTH responses. The T cell compartment of the spleen was capable of expressing this suppression. CD8(+) Tregs could solely express this CII-driven suppression and even exerted more noticeable suppression than spleen cells or splenic T cells. This study suggests a crucial role for CD8(+) Tregs in mediating CII-driven ACAID-mediated immune tolerance. This could have therapeutic implications in Rheumatoid arthritis, aging, osteoarthritis, and other diseases in which CII is involved.
Collapse
|
20
|
Reekmans K, Praet J, De Vocht N, Daans J, Van der Linden A, Berneman Z, Ponsaerts P. Stem cell therapy for multiple sclerosis: preclinical evidence beyond all doubt? Regen Med 2012; 7:245-59. [PMID: 22397612 DOI: 10.2217/rme.12.5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Stem cell transplantation holds great promise for restoration of neural function in various neurodegenerative disorders, including multiple sclerosis (MS). However, many questions remain regarding the true efficacy and precise mode of action of stem cell-based therapeutic approaches. Therefore, in this article, we will first discuss the ideal route and/or timing of stem cell-based therapies for experimental autoimmune encephalomyelitis (EAE), the most used preclinical animal model for MS. Next, we will provide an overview of the proposed mechanisms that contribute to the beneficial effects of stem cell transplantation observed during the treatment of rodent EAE. Reviews of current and past literature clearly demonstrate conceptual changes in the development of stem cell-based approaches for EAE/MS, leading to the identification of several major challenges to be tackled before (stem) cell therapy for rodent EAE can be safely and successfully translated to human therapy for MS.
Collapse
Affiliation(s)
- Kristien Reekmans
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (Vaxinfectio), University of Antwerp, Universiteitsplein 1, 2610 Antwerp, Belgium
| | | | | | | | | | | | | |
Collapse
|
21
|
Engels B, Rowley DA, Schreiber H. Targeting stroma to treat cancers. Semin Cancer Biol 2011; 22:41-9. [PMID: 22212863 DOI: 10.1016/j.semcancer.2011.12.008] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2011] [Accepted: 12/15/2011] [Indexed: 01/29/2023]
Abstract
All cancers depend on stroma for support of growth. Leukemias, solid tumors, cancer cells causing effusions, metastases as well as micro-disseminated cancer cells release factors that stimulate stromal cells, which in turn produce ligands that stimulate cancer cells. Therefore, elimination of stromal support by destroying the stromal cells or by inhibiting feedback stimulation of cancer growth is in the focus of many evolving therapies. A stringent evaluation of the efficacy of stromal targeting requires testing in animal models. Most current studies emphasize the successes of stromal targeting rather than deciphering its limitations. Here we show that many of the stromal targeting approaches, while often reducing tumor growth rates, are rarely curative. Therefore, we will also discuss conditions where stromal targeting can eradicate large established tumors. Finally, we will examine still unanswered questions of this promising and exciting area of cancer research.
Collapse
Affiliation(s)
- Boris Engels
- Department of Pathology, Committee on Cancer Biology, Committee on Immunology, The University of Chicago, Chicago, IL 60637-5420, USA.
| | | | | |
Collapse
|
22
|
Foureau DM, McKillop IH, Jones CP, Amin A, White RL, Salo JC. Skin tumor responsiveness to interleukin-2 treatment and CD8 Foxp3+ T cell expansion in an immunocompetent mouse model. Cancer Immunol Immunother 2011; 60:1347-56. [PMID: 21638127 PMCID: PMC11029730 DOI: 10.1007/s00262-011-1035-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2010] [Accepted: 05/10/2011] [Indexed: 12/23/2022]
Abstract
Recombinant human interleukin-2 (rhIL-2) therapy is approved for treating patients with advanced melanoma yet significant responses are observed in only 10-15% of patients. Interleukin-2 induces Foxp3 expression in activated human CD8 T cells in vitro and expands circulating CD8 Foxp3+ T cells in melanoma patients. Employing IL-2 responsive (B16-F1, B16-BL6, JB/MS, MCA-205) and nonresponsive (JB/RH, B16-F10) subcutaneous tumor mouse models, we evaluated CD8 Foxp3+ T cell distribution and changes in response to rhIL-2 (50,000 U, i.p. or s.q., twice daily for 5 days). In tumor-free mice and subcutaneous tumor-bearing mouse models, CD8 Foxp3+ T cells were a rare but naturally occurring cell subset. Primarily located in skin-draining lymph nodes, CD8 Foxp3+ T cells expressed both activated T cell (CD28(+), CD44(+)) and Treg (CTLA4(+), PD1(lo/var), NKG2A(+/var)) markers. Following treatment with rhIL-2, a dramatic increase in CD8 Foxp3+ T cell prevalence was observed in the circulation and tumor-draining lymph nodes (TD.LNs) of animals bearing IL-2 nonresponsive tumors, while no significant changes were observed in the circulation and TD.LNs of animals bearing IL-2 responsive tumors. These findings suggest expansion of CD8 Foxp3+ T cell population in response to rhIL-2 treatment may serve as an early marker for tumor responsiveness to immunotherapy in an immune competent model. Additionally, these data may provide insight to predict response in patients with melanoma undergoing rhIL-2 treatment.
Collapse
Affiliation(s)
- David M. Foureau
- Department of General Surgery, Carolinas Medical Center, 1000 Blythe Boulevard, Charlotte, NC 28203 USA
- Blumenthal Cancer Center, Carolinas Medical Center, 1000 Blythe Boulevard, Charlotte, NC 28203 USA
| | - Iain H. McKillop
- Department of General Surgery, Carolinas Medical Center, 1000 Blythe Boulevard, Charlotte, NC 28203 USA
| | - Chase P. Jones
- Department of General Surgery, Carolinas Medical Center, 1000 Blythe Boulevard, Charlotte, NC 28203 USA
| | - Asim Amin
- Blumenthal Cancer Center, Carolinas Medical Center, 1000 Blythe Boulevard, Charlotte, NC 28203 USA
- Department of Medicine, Carolinas Medical Center, 1000 Blythe Boulevard, Charlotte, NC 28203 USA
| | - Richard L. White
- Department of General Surgery, Carolinas Medical Center, 1000 Blythe Boulevard, Charlotte, NC 28203 USA
- Blumenthal Cancer Center, Carolinas Medical Center, 1000 Blythe Boulevard, Charlotte, NC 28203 USA
| | - Jonathan C. Salo
- Department of General Surgery, Carolinas Medical Center, 1000 Blythe Boulevard, Charlotte, NC 28203 USA
- Blumenthal Cancer Center, Carolinas Medical Center, 1000 Blythe Boulevard, Charlotte, NC 28203 USA
| |
Collapse
|
23
|
Current state of type 1 diabetes immunotherapy: incremental advances, huge leaps, or more of the same? Clin Dev Immunol 2011; 2011:432016. [PMID: 21785616 PMCID: PMC3139873 DOI: 10.1155/2011/432016] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2011] [Accepted: 04/28/2011] [Indexed: 01/09/2023]
Abstract
Thus far, none of the preclinically successful and promising immunomodulatory agents for type 1 diabetes mellitus (T1DM) has conferred stable, long-term insulin independence to diabetic patients. The majority of these immunomodulators are humanised antibodies that target immune cells or cytokines. These as well as fusion proteins and inhibitor proteins all share varying adverse event occurrence and severity. Other approaches have included intact putative autoantigens or autoantigen peptides. Considerable logistical outlays have been deployed to develop and to translate humanised antibodies targeting immune cells, cytokines, and cytokine receptors to the clinic. Very recent phase III trials with the leading agent, a humanised anti-CD3 antibody, call into question whether further development of these biologics represents a step forward or more of the same. Combination therapies of one or more of these humanised antibodies are also being considered, and they face identical, if not more serious, impediments and safety issues. This paper will highlight the preclinical successes and the excitement generated by phase II trials while offering alternative possibilities and new translational avenues that can be explored given the very recent disappointment in leading agents in more advanced clinical trials.
Collapse
|
24
|
Haroon F, Drögemüller K, Händel U, Brunn A, Reinhold D, Nishanth G, Mueller W, Trautwein C, Ernst M, Deckert M, Schlüter D. Gp130-Dependent Astrocytic Survival Is Critical for the Control of Autoimmune Central Nervous System Inflammation. THE JOURNAL OF IMMUNOLOGY 2011; 186:6521-31. [DOI: 10.4049/jimmunol.1001135] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
25
|
Jung S, Park YK, Shin JH, Lee H, Kim SY, Lee GR, Park SH. The requirement of natural killer T-cells in tolerogenic APCs-mediated suppression of collagen-induced arthritis. Exp Mol Med 2011; 42:547-54. [PMID: 20610917 DOI: 10.3858/emm.2010.42.8.055] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
TGF-beta-induced tolerogenic-antigen presenting cells (Tol-APCs) could induce suppression of autoimmune diseases such as collagen-induced arthritis (CIA) and allergic asthma. In contrast, many studies have shown that NKT cells are involved in the pathogenesis of Th1-mediated autoimmune joint inflammation and Th2-mediated allergic pulmonary inflammation. In this study, we investigated the effect of CD1d-restricted NKT cells in the Tol-APCs-mediated suppression of autoimmune disease using a murine CIA model. When CIA-induced mice were treated with Tol-APCs obtained from CD1d+/- or CD1d-/- mice, unlike CD1d+/- APCs, CD1d-/- Tol-APCs failed to suppress CIA. More specifically, CD1d-/- Tol-APCs failed to suppress the production of inflammatory cytokines and the induction of Th2 responses by antigen-specific CD4 T cells both in vitro and in vivo. Our results demonstrate that the presence of CD1d-restricted NKT cells is critical for the induction of Tol-APCs-mediated suppression of CIA.
Collapse
Affiliation(s)
- Sundo Jung
- School of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| | | | | | | | | | | | | |
Collapse
|
26
|
Sharabi A, Mozes E. Harnessing regulatory T cells for the therapy of lupus and other autoimmune diseases. Immunotherapy 2011; 1:385-401. [PMID: 20635958 DOI: 10.2217/imt.09.2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Regulatory T cells (Tregs) maintain immunological homeostasis and prevent autoimmunity. The depletion or functional alteration of Tregs may lead to the development of autoimmune diseases. Tregs consist of different subpopulations of cells, of which CD4(+)CD25(+)Foxp3(+) cells are the most well characterized. However, CD8 Tregs also constitute a major cell population that has been shown to play an important role in autoimmune diseases. This review will discuss the role of Tregs in autoimmune diseases in general and specifically in systemic lupus erythematosus (SLE). SLE is a multisystem autoimmune disease characterized by the production of autoantibodies against nuclear components and by the deposition of immune complexes in the kidneys as well as in other organs. Abnormalities in Tregs were reported in SLE patients and in animal models of the disease. Current treatment of SLE is based on immunosuppressive drugs that are nonspecific and may cause adverse effects. Therefore, the development of novel, specific, side effect-free therapeutic means that will induce functional Tregs is a most desirable goal. Our group and others have designed and utilized tolerogenic peptides that ameliorate SLE manifestations in murine models. Here, we demonstrate the role of CD4 and CD8 Tregs, as well as the interaction between the two subsets of cells and the mechanism of action of the tolerogenic peptides. We also discuss their therapeutic potential for the treatment of SLE.
Collapse
Affiliation(s)
- Amir Sharabi
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | | |
Collapse
|
27
|
Mantel PY, Schmidt-Weber CB. Transforming growth factor-beta: recent advances on its role in immune tolerance. Methods Mol Biol 2011; 677:303-38. [PMID: 20941619 DOI: 10.1007/978-1-60761-869-0_21] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Transforming growth factor (TGF-β1) is a pleiotropic cytokine, secreted by immune and nonhematopoietic cells. TGF-β is involved in many different critical processes, such as embryonal development, cellular maturation and differentiation, wound healing, and immune regulation. It maintains immune homeostasis by acting as a potent immune suppressor through inhibition of proliferation, differentiation, activation, and effector function of immune cells. Paradoxically, depending on the context, it displays proinflammatory properties by being a potent chemoattractant for neutrophils and promoting inflammation. In addition, it does not only induce differentiation into the anti-inflammatory Treg cells, but also into the proinflammatory Th17 and Th9 cells and inhibits Th22 differentiation. TGF-β has been demonstrated to be involved in multiple pathologies. In infections, it protects against collateral damages caused by the immune system, but it also promotes immune evasion and chronic infections. In autoimmune diseases, a TGF-β dysfunction leads to the loss of tolerance to self-antigens. In cancer, TGF-β is a potent inhibitor of cell proliferation and acts as a tumor suppressor at the beginning of tumorogenesis. However, once the cells become resistant to TGF-β, it mainly supports tumor growth and metastasis by promoting immune evasion and angiogenesis. In asthma, it is assumed to promote allergen tolerance, but plays a detrimental role in irreversible remodeling of the airways. Despite the high numbers of TGF-β-targeted pathways, it is a promising drug target for treatment of autoimmunity, cancer, fibrosis, if cell specificity can be achieved.This review summarizes the progresses that have been accomplished on the understanding of TGF-β's signaling in the immune homeostasis and its role in pathogenesis.
Collapse
Affiliation(s)
- Pierre-Yves Mantel
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Harvard University, Boston, MA, USA.
| | | |
Collapse
|
28
|
Bhowmick S, Clark RB, Brocke S, Cone RE. Antigen-specific splenic CD4+ and CD8+ regulatory T cells generated via the eye, suppress experimental autoimmune encephalomyelitis either at the priming or at the effector phase. Int Immunol 2011; 23:119-28. [PMID: 21273399 DOI: 10.1093/intimm/dxq461] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The injection of antigen into the ocular anterior chamber (AC) induces the generation of splenic CD4(+) and CD8(+) regulatory T (Treg) cells, specific for the antigen injected into the AC. These Treg cells inhibit the induction (CD4(+)) and also the expression (CD8(+)) of a delayed-type hypersensitivity response. The ability of AC-induced self-antigen-specific Treg cells in modulating autoimmunity is not well defined. Here we show that an injection of encephalitogenic myelin oligodendrocyte glycoprotein (MOG(35-55)) peptide into the anterior chamber of the eye (AC-MOG), before the induction of or during established experimental autoimmune encephalomyelitis (EAE) induced by MOG(35-55), suppresses the induction or progression of EAE, respectively. CD4(+) or CD8(+) splenic Treg cells induced by an injection of AC-MOG prevent EAE either at the inductive (priming) or at the progressive (effector) phase, respectively. This suppression of EAE by an AC-MOG injection or by intravenous transfer of splenic regulatory cells induced by an AC-MOG injection is specific for the antigen injected into the AC. Additionally, our data suggest that splenic CD8(+) Treg cells that suppress active EAE may use a transforming growth factor (TGF)-β-dependent suppression mechanism while the suppression of the induction of EAE by the AC-induced CD4(+) Treg cells is independent of TGF-β. Thus, we show for the first time that regulation of EAE at the priming or the chronic phase requires different phenotypes of Treg cells. Hence, it is important to consider the phenotype of Treg cells while designing effective cell-based therapies against autoimmune disorders.
Collapse
Affiliation(s)
- Sourojit Bhowmick
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06032-3105, USA
| | | | | | | |
Collapse
|
29
|
Wölfle SJ, Strebovsky J, Bartz H, Sähr A, Arnold C, Kaiser C, Dalpke AH, Heeg K. PD-L1 expression on tolerogenic APCs is controlled by STAT-3. Eur J Immunol 2011; 41:413-24. [PMID: 21268011 DOI: 10.1002/eji.201040979] [Citation(s) in RCA: 263] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2010] [Revised: 11/04/2010] [Accepted: 11/25/2010] [Indexed: 12/18/2022]
Abstract
During infection, TLR agonists are released and trigger mature as well as differentiating innate immune cells. Early encounter with TLR agonists (R848; LPS) blocks conventional differentiation of CD14(+) monocytes into immature dendritic cells (iDCs) resulting in a deviated phenotype. We and others characterized these APCs (TLR-APC) by a retained expression of CD14 and a lack of CD1a. Here, we show in addition, expression of programmed death ligand-1 (PD-L1). TLR-APCs failed to induce T-cell proliferation and furthermore were able to induce CD25(+) Foxp3(+) T regulatory cells (Tregs). Since PD-L1 is described as a key negative regulator and inducer of tolerance, we further analyzed its regulation. PD-L1 expression was regulated in a MAPK/cytokine/STAT-3-dependent manner: high levels of IL-6 and IL-10 that signal via STAT-3 were produced by TLR-APCs. Blocking of STAT-3 activation prevented PD-L1 expression. Moreover, chromatin immunoprecipitation revealed direct binding of STAT-3 to the PD-L1 promoter. Those findings indicate a pivotal role of STAT-3 in regulating PD-L1 expression. MAPKs were indirectly engaged, as blocking of p38 and p44/42 MAPKs decreased IL-6 and IL-10 thus reducing STAT-3 activation and subsequent PD-L1 expression. Hence, during DC differentiation TLR agonists induce a STAT-3-mediated expression of PD-L1 and favor the development of tolerogenic APCs.
Collapse
Affiliation(s)
- Sabine J Wölfle
- Department for Infectious Diseases, Medical Microbiology and Hygiene, University of Heidelberg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Švajger U, Obermajer N, Jeras M. Novel Findings in Drug-Induced Dendritic Cell Tolerogenicity. Int Rev Immunol 2010; 29:574-607. [DOI: 10.3109/08830185.2010.522280] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
31
|
Jung S, Park YK, Lee H, Shin JH, Lee GR, Park SH. TGF-beta-treated antigen presenting cells suppress collagen- induced arthritis through the promotion of Th2 responses. Exp Mol Med 2010; 42:187-94. [PMID: 20164680 DOI: 10.3858/emm.2010.42.3.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Collagen-induced arthritis (CIA) is mediated by self-reactive CD4(+) T cells that produce inflammatory cytokines. TGF-beta(2)-treated tolerogenic antigen-presenting cells (Tol-APCs) are known to induce tolerance in various autoimmune diseases. In this study, we investigated whether collagen-specific Tol-APCs could induce suppression of CIA. We observed that Tol-APCs could suppress the development and severity of CIA and delay the onset of CIA. Treatment of Tol-APCs reduced the number of IFN-gamma- and IL-17-producing CD4(+) T cells and increased IL-4- and IL-5-producing CD4(+) T cells upon collagen antigen stimulation in vitro. The suppression of CIA conferred by Tol-APCs correlated with their ability to selectively induce IL-10 production. We also observed that treatment of Tol-APCs inhibited not only cellular immune responses but also humoral immune responses in the process of CIA. Our results suggest that in vitro-generated Tol-APCs have potential therapeutic value for the treatment of rheumatoid arthritis as well as other autoimmune diseases.
Collapse
Affiliation(s)
- Sundo Jung
- School of Life Sciences and Biotechnology, Korea University, Seoul 136-701, Korea
| | | | | | | | | | | |
Collapse
|
32
|
Abstract
BACKGROUND The novel immunosuppressive molecule, CD200, has been reported to induce immunoregulation after interaction with its receptor(s), CD200R(s), in part at least through augmented induction of regulatory T-cell populations. Independent studies have also described increased expression of indoleamine-2,3-dioxygenase after CD200R triggering, whereas others have provided evidence that TGF-beta is important for the induction or function of many populations of regulatory T cells. We have asked whether a hybrid molecule in which a soluble fusion protein containing CD200, CD200Fc, was linked to TGF-beta through a glycine linker (Gly6) functions as a superior immunosuppressant molecule when compared with CD200Fc or TGF-beta alone, or in combination. METHODS The hybrid molecule CD200FcGly6TGF-beta was expressed by transient transfection in CHO cells and purified over a protein A column. Functional activity of this and recombinant CD200Fc or TGF-beta alone were assessed in mixed leukocyte cultures (MLCs) and in skin graft rejection in vivo. RESULTS Immunosuppression mediated by CD200FcGly6TGF-beta is dependent on both functional CD200 and TGF-beta moieties, as indicated by inhibition of suppression using anti-CD200 or anti-TGF-beta antibodies. Using as responder cells, using antigen-presenting cell from mice with a deletion of the CD200R gene and responder T cells from mice with siRNA-mediated suppression of expression of the TGF-betaII receptor, we show that suppression follows binding to TGF-betaRII on T cells, and CD200R1 on antigen-presenting cells. Indoleamine-2,3-dioxygenase inhibitors did not attenuate suppression by CD200FcGly6TGF-beta. CONCLUSION CD200FcGly6TGF-beta is a potent immunosuppressant in vivo and in vitro.
Collapse
|
33
|
Phillips B, Giannoukakis N, Trucco M. Dendritic cell-based therapy in Type 1 diabetes mellitus. Expert Rev Clin Immunol 2010; 5:325-39. [PMID: 20477010 DOI: 10.1586/eci.09.8] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Dendritic cell (DC) immunotherapy is a clinical reality. Despite two decades of considerable data demonstrating the feasibility of using DCs to prolong transplant allograft survival and to prevent autoimmunity, only now are these cells entering clinical trials in humans. Type 1 diabetes is the first autoimmune disorder to be targeted for treatment in humans using autologous-engineered DCs. This review will highlight the role of DCs in autoimmunity and the manner in which they have been engineered to treat these disorders in rodent models, either via the induction of immune hyporesponsiveness, which may be cell- and/or antigen-specific, or indirectly by upregulation of other immune cell networks.
Collapse
Affiliation(s)
- Brett Phillips
- University of Pittsburgh School of Medicine, Department of Pediatrics, Division of Immunogenetics, Children's Hospital of Pittsburgh, Rangos Research Center, 530 45th Street, Pittsburgh, PA 15201, USA.
| | | | | |
Collapse
|
34
|
Svajger U, Obermajer N, Jeras M. Dendritic cells treated with resveratrol during differentiation from monocytes gain substantial tolerogenic properties upon activation. Immunology 2010; 129:525-35. [PMID: 20002210 PMCID: PMC2842499 DOI: 10.1111/j.1365-2567.2009.03205.x] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2009] [Revised: 10/20/2009] [Accepted: 10/21/2009] [Indexed: 12/13/2022] Open
Abstract
Resveratrol is a polyphenol that acts on multiple molecular targets important for cell differentiation and activation. Dendritic cells (DCs) are a functionally diverse cell type and represent the most potent antigen-presenting cells of the immune system. In this study, we investigated resveratrol-induced effects on DCs during their differentiation and maturation. Our results show that resveratrol induces DC-associated tolerance, particularly when applied during DC differentiation. Costimulatory molecules CD40, CD80 and CD86 were down-regulated, as was the expression of major histocompatibility complex (MHC) class II molecules. Surface expression of inhibitory immunoglobulin-like transcript 3 (ILT3) and ILT4 molecules was induced, while human leucocyte antigen (HLA)-G expression was not affected. Resveratrol-treated DCs lost the ability to produce interleukin (IL)-12p70 after activation, but had an increased ability to produce IL-10. Such DCs were poor stimulators of allogeneic T cells and had lowered ability to induce CD4(+) T-cell migration. Furthermore, treated cells were able to generate allogeneic IL-10-secreting T cells, but were not competent in inducing FoxP3 expression These tolerogenic effects are probably associated with the effect of resveratrol on multiple molecular targets through which it interferes with DC differentiation and nuclear factor (NF)-kappaB translocation. Our data provide new insights into the molecular and functional mechanisms of the tolerogenic effects that resveratrol exerts on DCs.
Collapse
Affiliation(s)
- Urban Svajger
- Blood Transfusion Center of Slovenia, Slajmerjeva 6, Ljubljana, Slovenia.
| | | | | |
Collapse
|
35
|
Abstract
Langerhans cells (LC) are members of the heterogenous family of professional antigen presenting dendritic cells (DC). They are identified by the C-type lectin receptor Langerin and form a contiguous network in the epidermis. Consequently, LC are an integral part of the skin barrier to the environment and were considered to be critical inducers of skin immunity, whereas dermal DC were largely overlooked. However, with the identification of a distinct subset of Langerin expressing dermal DC, the situation in the skin has become more complex and the relative contribution of the different cutaneous DC populations in balancing immunity and tolerance has become a matter of active debate. Here, we briefly review the classical paradigm and recent challenges of LC function, before focusing on advances concerning their role in contact hypersensitivity and ultraviolet radiation-induced immunosuppression obtained with in vivo LC ablation models. We then discuss novel LC/DC-specific gene targeting approaches currently used to dissect the role of the regulatory cytokines transforming growth factor-beta and interleukin-10 to govern LC and DC function in vivo. This second generation of LC-specific genetically engineered mice will considerably extend our understanding of the molecular control of LC function in regulating skin immunity and tolerance in the near future.
Collapse
Affiliation(s)
- Björn E Clausen
- Department of Immunology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands.
| | | |
Collapse
|
36
|
Wolf B, Posnick D, Fisher JL, Lewis LD, Ernstoff MS. Indoleamine-2,3-dioxygenase enzyme expression and activity in polarized dendritic cells. Cytotherapy 2010; 11:1084-9. [PMID: 19929471 DOI: 10.3109/14653240903271230] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND AIMS Polarized mature dendritic cells (DC) can activate cytolytic T-lymphocyte (CTL) responses and may be a more effective clinical strategy in DC-based cancer vaccines. A subset of mature DC can down-regulate the T-cell immune response through expression of indoleamine-2,3-dioxygenase (IDO). We determined whether polarizing DC ex vivo increased IDO expression and activity. METHODS Peripheral blood monocytes from healthy volunteers were cultured ex vivo in polarizing and non-polarizing culture conditions. DC IDO expression was detected by Western blot. IDO enzyme activity was determined by high-performance liquid chromatography (HPLC) measurement of kynurenine (K) and tryptophan (T) concentrations in culture supernatants. RESULTS IDO protein was markedly increased in DC after polarization (median 1222.4%, range 331.5-2113.3%) versus non-polarized DC (median 28.3%, range 3.7-119.8%; P=0.04). The median K/T ratio was significantly higher in polarized DC versus non-polarized DC (6.34, range 6.02-6.65, versus 0.047, range 0.004-0.541; P=0.04). IDO protein expression correlated with enzyme activity (r=0.80, P=0.002). CONCLUSIONS DC polarizing culture conditions increased expression of IDO protein and IDO enzyme activity. DC culture and maturation methodologies may impact the effectiveness of adoptive DC therapy.
Collapse
Affiliation(s)
- Benita Wolf
- Medical Oncology Immunotherapy Group, Dartmouth Hitchcock Medical Center, Lebanon, New Hampshire 03756, USA
| | | | | | | | | |
Collapse
|
37
|
Zheng J, Liu Y, Qin G, Chan PL, Mao H, Lam KT, Lewis DB, Lau YL, Tu W. Efficient induction and expansion of human alloantigen-specific CD8 regulatory T cells from naive precursors by CD40-activated B cells. THE JOURNAL OF IMMUNOLOGY 2009; 183:3742-50. [PMID: 19684082 DOI: 10.4049/jimmunol.0901329] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Although recent studies have focused on CD4(+) regulatory T cells (Treg), CD8(+) Treg have also been reported to play important roles in the induction and maintenance of immune tolerance. Adoptive transfer of CD8(+) Treg in rodents or induction of CD8(+) Treg in humans can prevent or treat allograft rejection and autoimmune diseases. However, no approaches have been reported for the generation of human Ag-specific CD8(+) Treg at a practical scale for clinical use. Here, we found that two novel CD8(+) T cell subsets with different levels of CD8 surface expression, CD8(high) and CD8(low), could be induced from naive CD8(+) precursors in vitro by allogeneic CD40-activated B cells, whereas only CD8(high) T cells were alloantigen-specific Treg with relatively poor alloantigen-specific cytotoxicity. Importantly, alloantigen-specific CD8(high) Treg could be induced and expanded from naive CD8(+)CD25(-) T cells at a large scale after 3 wk of culture without exogenous cytokines. These induced alloantigen-specific Treg were CD45RO(+) and CCR7(-) memory cells, and they expressed Foxp3, CD25, CD27, CD28, and CD62L. The induction and expansion of CD8(high) Treg by CD40-activated B cells were dependent on endogenously expressed IFN-gamma, IL-2, IL-4, and CTLA-4. This approach may facilitate the clinical application of CD8(+) Treg-based immunotherapy in transplantation and autoimmune diseases.
Collapse
Affiliation(s)
- Jian Zheng
- Department of Pediatrics and Adolescent Medicine, LKS Faculty of Medicine, University of Hong Kong, Hong Kong SAR, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
Much emphasis has been placed on the so-called "biologics" in the treatment of immune disorders within the last few years. Here we discuss the expanding horizon of potential strategies for immunotherapies targeting T lymphocytes as key effectors and regulators of autoimmunity. We review emerging reagents in a variety of animal models and human disorders that may offer new therapeutic options in current or modified iterations.
Collapse
Affiliation(s)
- Erica Lee
- Department of Dermatology, Weill Medical College of Cornell University, New York, NY, USA
| | | |
Collapse
|
39
|
Abstract
Dendritic cells (DC) have been implicated both in initiation of immunity and in immune tolerance. The mechanisms whereby tolerogenic DC may induce and maintain peripheral tolerance include the generation or expansion of regulatory T cells (Treg) and the promotion of T-cell anergy or deletion. A wide spectrum of hematopoietic growth factors and cytokines are endowed with the ability to differentiate tolerogenic DC both in vitro and in vivo. Based on this knowledge, therapeutic vaccination with cytokine-modulated tolerogenic DC has been applied to animal models of autoimmune disorders. This article will review the current experimental evidence underpinning DC dysfunction in rheumatic autoimmune diseases and will discuss how the manipulation of DC and Treg number and function may control undesired T-cell responses.
Collapse
|
40
|
Masli S, Turpie B. Anti-inflammatory effects of tumour necrosis factor (TNF)-alpha are mediated via TNF-R2 (p75) in tolerogenic transforming growth factor-beta-treated antigen-presenting cells. Immunology 2009; 127:62-72. [PMID: 18795974 DOI: 10.1111/j.1365-2567.2008.02933.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Exposure of macrophages to transforming growth factor (TGF)-beta is known to alter their functional phenotype such that antigen presentation by these cells leads to tolerance rather than an inflammatory immune response. Typically, eye-derived antigen-presenting cells (APCs) exposed to TGF-beta in the local environment are known to induce a form of peripheral tolerance and protect the eye from inflammatory immune effector-mediated damage. In response to TGF-beta, APCs increase their expression of tumour necrosis factor (TNF)-alpha and TNF receptor 2 (TNF-R2). Although TNF-alpha has been implicated in tolerance and the associated regulation of the inflammatory immune response, its source and the receptors involved remain unclear. In this report we determined the contribution of TNF-alpha and TNF-R2 expressed by TGF-beta-treated APCs to their anti-inflammatory tolerogenic effect. Our results indicate that APC-derived TNF-alpha is essential for the ability of APCs to regulate the immune response and their IL-12 secretion. Moreover, in the absence of TNF-R2, APCs exposed to TGF-beta failed to induce tolerance or regulatory cells known to participate in this tolerance. Also, blocking of TNF-R1 signalling enhanced the ability of the APCs to secrete increased TGF-beta in response to TGF-beta exposure. Together our results support an anti-inflammatory role of TNF-alpha in regulation of an immune response by TGF-beta-treated APCs and suggest that TNF-R2 contributes significantly to this role.
Collapse
Affiliation(s)
- Sharmila Masli
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA.
| | | |
Collapse
|
41
|
Cui Y, Shao H, Sun D, Kaplan HJ. Regulation of interphotoreceptor retinoid-binding protein (IRBP)-specific Th1 and Th17 cells in anterior chamber-associated immune deviation (ACAID). Invest Ophthalmol Vis Sci 2009; 50:5811-7. [PMID: 19458338 DOI: 10.1167/iovs.09-3389] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
PURPOSE Intracameral (anterior chamber) injection of antigen inhibits the development of delayed-type hypersensitivity, a phenomenon known as anterior chamber-associated immune deviation (ACAID). The authors investigated the effect of intracameral injection of interphotoreceptor retinoid-binding protein (IRPB) peptides on the development of IFN-gamma(+) and IL-17(+) pathogenic T cells. METHODS A uveitogenic (IRBP1-20) or nonuveitogenic (IRBP161-180) peptide was injected into the anterior chamber (AC) of B6 mice. Seven days later, the mice were primed with a pathogenic dose of IRBP1-20 in adjuvant. Thirteen days later, the pathogenic activity of the T cells isolated from the spleens of treated and untreated mice were compared, and the numbers of Th1 and Th17 T cells were assessed by intracellular staining. Regulatory T-cell activity was assessed by antibody staining and functional assays. The authors also compared the effect of inhibition on EAU of ocular injection to various sites, including the AC, the vitreous cavity, and the subretinal space. RESULTS Intraocular injection of the uveitogenic peptide (IRBP1-20), but not the nonuveitogenic peptide (IRBP161-180), inhibited the generation of IFN-gamma(+) and IL-17(+) uveitogenic T cells and the development of experimental autoimmune uveitis (EAU). AC administration of IRBP1-20, but not IRBP161-180, significantly decreased the number of circulating gammadelta T cells after subsequent systemic immunization with IRBP1-20. Absence of the gammadelta T-cell population prohibited the development of ACAID. CONCLUSIONS Injection of a uveitogenic peptide into the AC inhibited the development of EAU by regulation of Th1 and Th17 IRBP-specific T cells. The circulating gammadelta T-cell population was reduced and was associated with decreased activation of IL-17(+) uveitogenic T cells.
Collapse
Affiliation(s)
- Yan Cui
- Department of Ophthalmology and Visual Sciences, Kentucky Lions Eye Center, University of Louisville, Louisville, Kentucky 40202, USA
| | | | | | | |
Collapse
|
42
|
Cone RE, Chattopadhyay S, Sharafieh R, Lemire Y, O'Rourke J, Flavell RA, Clark RB. T cell sensitivity to TGF-beta is required for the effector function but not the generation of splenic CD8+ regulatory T cells induced via the injection of antigen into the anterior chamber. Int Immunol 2009; 21:567-74. [PMID: 19325036 PMCID: PMC2675031 DOI: 10.1093/intimm/dxp023] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The introduction of antigen into the anterior chamber (AC) of the eye induces the production of antigen-specific splenic CD8+ regulatory T cells (AC-SPL cells) that suppress a delayed-type hypersensitivity (DTH) reaction in immunized mice. Because the generation of these regulatory T cells is also induced by exposure to transforming growth factor (TGF)-β and antigen or F4/80+ cells exposed to TGF-β and antigen in vitro, we investigated (i) whether these cells are produced in dominant negative receptor for transforming growth factor β receptor type II (dnTGFβRII) or Cbl-b−/− mice whose T cells are resistant to TGF-β, (ii) whether DTH is suppressed by wild type (WT) CD8+ AC-SPL cells in Cbl-b−/− and dnTGFβRII mice and (iii) the effect of antibodies to TGF-β on the suppression of DTH by CD8+ AC-SPL cells. DnTGFβRII immunized and Cbl-b−/− mice produced splenic CD8+ regulatory cells after the intracameral injection of antigen and immunization. The suppression of a DTH reaction by CD8+ AC-SPL cells in WT mice was blocked by the local inclusion of antibodies to TGF-β when WT splenic CD8+ AC-SPL cells were injected into the DTH reaction site. Moreover, the DTH reaction in immunized dnTGFβRII and Cbl-b−/− mice was not suppressed by the transfer of WT CD8+ AC-SPL cells to the site challenged with antigen. In aggregate, these observations suggest that T cell sensitivity to TGF-β is not an obligate requirement for the in vivo induction of CD8+ AC-SPL T cells but the suppression of an in vivo DTH reaction by CD8+ AC-SPL cells is dependent on TGF-β.
Collapse
Affiliation(s)
- Robert E Cone
- Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030-3105, USA.
| | | | | | | | | | | | | |
Collapse
|
43
|
Fudaba Y, Onoe T, Chittenden M, Shimizu A, Shaffer JM, Bronson R, Sykes M. Abnormal regulatory and effector T cell function predispose to autoimmunity following xenogeneic thymic transplantation. THE JOURNAL OF IMMUNOLOGY 2008; 181:7649-59. [PMID: 19017953 DOI: 10.4049/jimmunol.181.11.7649] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Porcine thymus grafts support robust murine and human thymopoiesis, generating a diverse T cell repertoire that is deleted of donor and host-reactive cells, achieving specific xenograft tolerance. Positive selection is mediated exclusively by the xenogeneic thymic MHC. Although thymectomized, T cell-depleted normal mice usually remain healthy following xenogeneic thymic transplantation, thymus-grafted congenitally athymic mice frequently develop multiorgan autoimmunity. We investigated the etiology of this syndrome by adoptively transferring lymphocyte populations from fetal pig thymus-grafted BALB/c nude mice to secondary BALB/c nude recipients. Fetal pig thymus-grafted nude mice generated normal numbers of CD25(+)Foxp3(+)CD4 T cells, but these cells lacked the capacity to block autoimmunity. Moreover, thymocytes and peripheral CD4(+)CD25(-) cells from fetal pig thymus-grafted nude mice, but not those from normal mice, induced autoimmunity in nude recipients. Injection of thymic epithelial cells from normal BALB/c mice into fetal pig thymus grafts reduced autoimmunity and enhanced regulatory function of splenocytes. Our data implicate abnormalities in postthymic maturation, expansion, and/or survival of T cells positively selected by a xenogeneic MHC, as well as incomplete intrathymic deletion of thymocytes recognizing host tissue-specific Ags, in autoimmune pathogenesis. Regulatory cell function is enhanced and negative selection of host-specific thymocytes may potentially also be improved by coimplantation of recipient thymic epithelial cells in the thymus xenograft.
Collapse
Affiliation(s)
- Yasuhiro Fudaba
- Department of Surgery, Transplantation Biology Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA 02129, USA
| | | | | | | | | | | | | |
Collapse
|
44
|
Li Q, Guo Z, Xu X, Xia S, Cao X. Pulmonary stromal cells induce the generation of regulatory DC attenuating T-cell-mediated lung inflammation. Eur J Immunol 2008; 38:2751-61. [PMID: 18825748 DOI: 10.1002/eji.200838542] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The tissue microenvironment may affect the development and function of immune cells such as DC. Whether and how the pulmonary stromal microenvironment can affect the development and function of lung DC need to be investigated. Regulatory DC (DCreg) can regulate T-cell response. We wondered whether such regulatory DC exist in the lung and what is the effect of the pulmonary stromal microenvironment on the generation of DCreg. Here we demonstrate that murine pulmonary stromal cells can drive immature DC, which are regarded as being widely distributed in the lung, to proliferate and differentiate into a distinct subset of DCreg, which express high levels of CD11b but low levels of MHC class II (I-A), CD11c, secrete high amounts of IL-10, NO and prostaglandin E2 (PGE2) and suppress T-cell proliferation. The natural counterpart of DCreg in the lung with similar phenotype and regulatory function has been identified. Pulmonary stroma-derived TGF-beta is responsible for the differentiation of immature DC to DCreg, and DCreg-derived PGE2 contributes to their suppression of T-cell proliferation. Moreover, DCreg can induce the generation of CD4+CD25+Foxp3+ Treg. Importantly, infusion with DCreg attenuates T-cell-mediated eosinophilic airway inflammation in vivo. Therefore, the pulmonary microenvironment may drive the generation of DCreg, thus contributing to the maintenance of immune homoeostasis and the control of inflammation in the lung.
Collapse
Affiliation(s)
- Qian Li
- Institute of Immunology and National Key Laboratory of Medical Immunology, Second Military Medical University, Shanghai, P. R. China
| | | | | | | | | |
Collapse
|
45
|
Jarvis LB, Goodall JC, Gaston JH. Human leukocyte antigen class I-restricted immunosuppression by human CD8+ regulatory T cells requires CTLA-4-mediated interaction with dendritic cells. Hum Immunol 2008; 69:687-95. [DOI: 10.1016/j.humimm.2008.08.277] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2008] [Revised: 08/11/2008] [Accepted: 08/12/2008] [Indexed: 12/29/2022]
|
46
|
McPherson M, Wei B, Turovskaya O, Fujiwara D, Brewer S, Braun J. Colitis immunoregulation by CD8+ T cell requires T cell cytotoxicity and B cell peptide antigen presentation. Am J Physiol Gastrointest Liver Physiol 2008; 295:G485-92. [PMID: 18617557 PMCID: PMC2536787 DOI: 10.1152/ajpgi.90221.2008] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Deficient immunoregulation by CD4+ T cells is an important susceptibility trait for inflammatory bowel disease, but the role of other regulatory cell types is less understood. This study addresses the role and mechanistic interaction of B cells and CD8+ T cells in controlling immune-mediated colitis. The genetic requirements for B cells and CD8+ T cells to confer protective immunoregulation were assessed by cotransfer with colitogenic Galphai2-/- T cells into immune-deficient mice. Disease activity in Galphai2-/- T cell recipients was evaluated by CD4+ T intestinal lymphocyte abundance, cytokine production levels, and large intestine histology. B cells deficient in B7.1/B7.2, CD40, major histocompatibility complex (MHC) II (Abb), or native B cell antigen receptor (MD4) were competent for colitis protection. However, transporter-1-deficient B cells failed to protect, indicating a requirement for peptide MHC I presentation to CD8+ T cells. CD8+ T cells deficient in native T cell receptor repertoire (OT-1) or cytolysis (perforin-/-) also were nonprotective. These finding reveal an integrated role for antigen-specific perforin-dependent CD8+ T cell cytotoxicity in colitis immunoregulatory and its efficient induction by a subset of mesenteric B lymphocytes.
Collapse
Affiliation(s)
- Michael McPherson
- Molecular Biology Institute, Department of Pathology and Laboratory Medicine, University of California, Los Angeles; and La Jolla Institute for Allergy and Immunology, San Diego, California
| | - Bo Wei
- Molecular Biology Institute, Department of Pathology and Laboratory Medicine, University of California, Los Angeles; and La Jolla Institute for Allergy and Immunology, San Diego, California
| | - Olga Turovskaya
- Molecular Biology Institute, Department of Pathology and Laboratory Medicine, University of California, Los Angeles; and La Jolla Institute for Allergy and Immunology, San Diego, California
| | - Daisuke Fujiwara
- Molecular Biology Institute, Department of Pathology and Laboratory Medicine, University of California, Los Angeles; and La Jolla Institute for Allergy and Immunology, San Diego, California
| | - Sarah Brewer
- Molecular Biology Institute, Department of Pathology and Laboratory Medicine, University of California, Los Angeles; and La Jolla Institute for Allergy and Immunology, San Diego, California
| | - Jonathan Braun
- Molecular Biology Institute, Department of Pathology and Laboratory Medicine, University of California, Los Angeles; and La Jolla Institute for Allergy and Immunology, San Diego, California
| |
Collapse
|
47
|
Lee VWS, Wang YM, Wang YP, Zheng D, Polhill T, Cao Q, Wu H, Alexander IE, Alexander SI, Harris DCH. Regulatory immune cells in kidney disease. Am J Physiol Renal Physiol 2008; 295:F335-42. [DOI: 10.1152/ajprenal.00077.2008] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Lymphocytes and macrophages act as effector immune cells in the initiation and progression of renal injury. Recent data have shown that subpopulations of these immune cells (regulatory T lymphocytes and alternately-activated or regulatory macrophages) are potent modulators of tissue injury and repair in renal disease. Recent animal studies examining the therapeutic effect of these cells raise the exciting possibility that strategies targeting these cell types may be effective in treating and preventing kidney disease in humans. This review will describe their biological role in experimental kidney disease and therapeutic potential in clinical nephrology.
Collapse
|
48
|
Katagiri K, Arakawa S, Hatano Y, Fujiwara S. Tolerogenic antigen-presenting cells successfully inhibit atopic dermatitis-like skin lesion induced by repeated epicutaneous exposure to ovalbumin. Arch Dermatol Res 2008; 300:583-93. [PMID: 18528699 DOI: 10.1007/s00403-008-0865-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2008] [Revised: 04/08/2008] [Accepted: 05/02/2008] [Indexed: 10/22/2022]
Abstract
Atopic dermatitis (AD) is a chronic pruritic inflammatory skin disease that frequently begins at infancy and the majority of them develop asthma and/or allergic rhinitis later, in which food and inhaled allergens play an important role. There is a murine model for AD that is induced by repeated epicutaneous (e.c.) exposure with ovalbumin (OVA). This model shares many characteristic features with AD, including development of asthma as well as dermatitis. Recently, it is reported that ocular tolerance or tolerance induced by intravenous administration of in vitro generated tolerogenic antigen-presenting cells (tol-APC), which can bypasses ocular tolerance, inhibits the immune response in a murine asthma model. The present study was designed to investigate whether tolerance induced by tol-APC and ocular tolerance inhibits AD-like dermatitis induced by repeated e.c. sensitization with OVA. BALB/c mice were given a total of three 1 week e.c. exposures to OVA with 2-week intervals between exposures. After second exposure to OVA, mice received the tol-APC or received OVA in the anterior chamber (AC) of the eye (ocular tolerance). Both groups of mice received the tol-APC and mice that received OVA in the AC of the eye showed weakened cellular infiltration in the skin including eosinophils and mast cells, lower levels of antigen-specific IgE, lower levels of transcripts of IL-4, IL-5, IL-13 in the skin and less production of Th1 and Th2 cytokine by regional lymph node cells, compared with those of mice that received sham treatment and mice that received the tol-APC treated with unrelated antigen after second e.c. exposure to OVA. These results indicate that antigen-specific tolerance induced by the tol-APC and ocular tolerance can inhibit the dermatitis and its related systemic immune response in the murine AD model. These types of tolerance might lead to a new therapeutic approach to allergic skin disease.
Collapse
Affiliation(s)
- Kazumoto Katagiri
- Department of Dermatology, Faculty of Medicine, Oita University, Idaigaoka 1-1, Hasama-machi, Oita 879-5593, Japan.
| | | | | | | |
Collapse
|
49
|
Abstract
The TNFR family members OX40 (CD134) and 4-1BB (CD137) have been found to play major roles as costimulatory receptors for both CD4 and CD8 T cells. In particular, in many situations, they can control proliferation, survival, and cytokine production, and hence are thought to dictate accumulation of protective T cells during anti-viral and anti-tumor responses and pathogenic T cells during autoimmune reactions. As opposed to simply controlling the activity of naïve, effector, and memory T cells, recent data have suggested that both molecules are also instrumental in controlling the generation and activity of so-called regulatory or suppressor T cells (Treg), perhaps in both positive and negative manners. Part of the action on Treg might function to further promote protective or pathogenic T cells, but alternate activities of OX40 and 4-1BB on Treg are also being described that suggest that there might be control by these molecules at multiple levels that will alter the biological outcome when these receptors are ligated. This review specifically focuses on recent studies of regulatory T cells, and regulatory or suppressive activity, that are modulated by OX40 or 4-1BB.
Collapse
Affiliation(s)
- Takanori So
- Division of Molecular Immunology, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | | | | |
Collapse
|
50
|
Smith TRF, Kumar V. Revival of CD8+ Treg-mediated suppression. Trends Immunol 2008; 29:337-42. [PMID: 18514574 DOI: 10.1016/j.it.2008.04.002] [Citation(s) in RCA: 158] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2008] [Revised: 04/18/2008] [Accepted: 04/18/2008] [Indexed: 01/19/2023]
Abstract
Despite their first recognition almost 40 years ago, CD8(+) 'suppressor' T cells remain poorly characterized. Recent studies of these lymphocytes, now popularly referred to as regulatory CD8(+) T cells (CD8(+) Tregs), have helped clarify their important role in the regulation of autoimmune disease. Here, we review progress related to the identification, phenotype and function of CD8(+) Tregs. We also focus on a newly described subset, CD8alphaalpha(+)TCRalphabeta(+) Tregs, which in mice recognize a T-cell receptor-derived peptide in the context of the class Ib major histocompatibility complex molecule Qa-1. These Tregs target only activated T cells and complement the suppression provided by CD4(+)Foxp3(+) Tregs. Investigations leading to the detailed identification, expansion, maintenance and function of CD8alphaalpha(+) Tregs should result in new therapeutic strategies for human inflammatory diseases.
Collapse
Affiliation(s)
- Trevor R F Smith
- Laboratory of Autoimmunity, Torrey Pines Institute for Molecular Studies, San Diego, CA 92121, USA
| | | |
Collapse
|