1
|
McCall JL, Blair HC, Blethen KE, Hall C, Elliott M, Barnett JB. Prenatal cadmium exposure does not induce greater incidence or earlier onset of autoimmunity in the offspring. PLoS One 2021; 16:e0249442. [PMID: 34478449 PMCID: PMC8415597 DOI: 10.1371/journal.pone.0249442] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 08/23/2021] [Indexed: 11/30/2022] Open
Abstract
We previously demonstrated that exposure of adult mice to environmental levels of cadmium (Cd) alters immune cell development and function with increases in anti-streptococcal antibody levels, as well as decreases in splenic natural regulatory T cells (nTreg) in the adult female offspring. Based on these data, we hypothesized that prenatal Cd exposure could predispose an individual to developing autoimmunity as adults. To test this hypothesis, the effects of prenatal Cd on the development of autoimmune diabetes and arthritis were investigated. Non-obese diabetic (NOD) mice were exposed to Cd in a manner identical to our previous studies, and the onset of diabetes was assessed in the offspring. Our results showed a similar time-to-onset and severity of disease to historical data, and there were no statistical differences between Cd-exposed and control offspring. Numerous other immune parameters were measured and none of these parameters showed biologically-relevant differences between Cd-exposed and control animals. To test whether prenatal Cd-exposure affected development of autoimmune arthritis, we used SKG mice. While the levels of arthritis were similar between Cd-exposed and control offspring of both sexes, the pathology of arthritis determined by micro-computed tomography (μCT) between Cd-exposed and control animals, showed some statistically different values, especially in the female offspring. However, the differences were small and thus, the biological significance of these changes is open to speculation. Overall, based on the results from two autoimmune models, we conclude that prenatal exposure to Cd did not lead to a measurable propensity to develop autoimmune disease later in life.
Collapse
Affiliation(s)
- Jamie L. McCall
- Department of Microbiology, Immunology & Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States of America
| | - Harry C. Blair
- Department of Pathology, Pittsburgh VA Medical Center, Pittsburgh, PA, United States of America
- Department of Cell Biology, the and the University of Pittsburgh, Pittsburgh, PA, United States of America
| | - Kathryn E. Blethen
- Department of Microbiology, Immunology & Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States of America
| | - Casey Hall
- Department of Microbiology, Immunology & Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States of America
| | - Meenal Elliott
- Department of Microbiology, Immunology & Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States of America
| | - John B. Barnett
- Department of Microbiology, Immunology & Cell Biology, West Virginia University School of Medicine, Morgantown, WV, United States of America
- West Virginia University Cancer Institute, West Virginia University, Morgantown, WV, United States of America
| |
Collapse
|
2
|
Hensvold A, Klareskog L. Towards prevention of autoimmune diseases: The example of rheumatoid arthritis. Eur J Immunol 2021; 51:1921-1933. [PMID: 34110013 DOI: 10.1002/eji.202048952] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 04/18/2021] [Indexed: 12/16/2022]
Abstract
Prevention is the ultimate aim for clinicians and scientists concerned with severe diseases, like many immune-mediated conditions. Here, we describe recent progress in the understanding of etiology and molecular pathogenesis of rheumatoid arthritis (RA), which make this disease a potential prototype for prevention that may include both public health measures and targeted and personalized approaches that we call "personalized prevention." Critical components of this knowledge are (i) better understanding of the dynamics of the RA-associated autoimmunity that may begin many years before onset of joint inflammation; (ii) insights into how this immunity may be triggered at mucosal surfaces after distinct environmental challenges; (iii) better understanding of which features of the pre-existing immunity may cause symptoms that precede joint inflammation and predict a high risk for imminent arthritis development; and (iv) how molecular events occurring before onset of inflammation might be targeted by existing or future therapies, ultimately by specific targeting of Major histocompatibility complex (MHC) class II restricted and RA-specific immunity. Our main conclusion is that studies and interventions in the phase of autoimmunity preceding RA offer new opportunities to prevent the disease and thereby also understand the molecular pathogenesis of its different variants.
Collapse
Affiliation(s)
- Aase Hensvold
- Division of Rheumatology, Department of Medicine, Karolinska Institutet and Karolinska University Hospital (Solna), Stockholm, Sweden.,Center for Rheumatology, Academic Specialist Center, Stockholm, Sweden
| | - Lars Klareskog
- Division of Rheumatology, Department of Medicine, Karolinska Institutet and Karolinska University Hospital (Solna), Stockholm, Sweden.,Center for Rheumatology, Academic Specialist Center, Stockholm, Sweden.,Rheumatology Section, Theme inflammation and Infection, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
3
|
Zhu J, Inomata T, Fujimoto K, Uchida K, Fujio K, Nagino K, Miura M, Negishi N, Okumura Y, Akasaki Y, Hirosawa K, Kuwahara M, Eguchi A, Shokirova H, Yanagawa A, Midorikawa-Inomata A, Murakami A. Ex Vivo-Induced Bone Marrow-Derived Myeloid Suppressor Cells Prevent Corneal Allograft Rejection in Mice. Invest Ophthalmol Vis Sci 2021; 62:3. [PMID: 34061951 PMCID: PMC8185403 DOI: 10.1167/iovs.62.7.3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 05/02/2021] [Indexed: 12/12/2022] Open
Abstract
Purpose To investigate the effects of ex vivo-induced bone marrow myeloid-derived suppressor cells (BM-MDSCs) on allogeneic immune responses in corneal transplantation. Methods Bone marrow cells from C57BL/6J (B6) mice were cultured with IL-6 and GM-CSF for four days. The ex vivo induction of the BM-MDSCs was assessed using flow cytometry, inducible nitric oxide synthase (iNOS) mRNA expression using reverse transcription-quantitative polymerase chain reaction, and nitric oxide (NO) production in allogeneic stimulation. T-cell proliferation and regulatory T-cell (Treg) expansion were investigated on allogeneic stimulation in the presence of ex vivo-induced BM-MDSCs. IFN-γ, IL-2, IL-10, and TGF-β1 protein levels were measured using enzyme-linked immunosorbent assays. After subconjunctival injection of ex vivo-induced BM-MDSCs, the migration of the BM-MDSCs into corneal grafts, allogeneic corneal graft survival, neovascularization, and lymphangiogenesis were assessed using flow cytometry, slit-lamp microscopy, and immunohistochemistry. Results The combination of GM-CSF and IL-6 significantly induced BM-MDSCs with increased iNos mRNA expression. The ex vivo-induced BM-MDSCs promoted NO release in allogeneic stimulation in vitro. The ex vivo-induced BM-MDSCs inhibited T-cell proliferation and promoted Treg expansion. Decreased IFN-γ and increased IL-2, IL-10, and TGF-β1 production was observed in coculture of ex vivo-induced BM-MDSCs. Injected ex vivo-induced BM-MDSCs were confirmed to migrate into the grafts. The injected BM-MDSCs also prolonged corneal graft survival and prevented angiogenesis and lymphangiogenesis. Conclusions The ex vivo-induced BM-MDSCs have suppressive effects on allogeneic immune responses and prolong corneal allograft survival via the iNOS pathway, indicating that they may be a potential therapeutic tool for corneal transplantation.
Collapse
Affiliation(s)
- Jun Zhu
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Ophthalmology, Subei People's Hospital Affiliated to Yangzhou University, Jiangsu Province, China
- Department of Ophthalmology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Takenori Inomata
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Ophthalmology, Juntendo University Faculty of Medicine, Tokyo, Japan
- Department of Strategic Operating Room Management and Improvement, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Hospital Administration, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Keiichi Fujimoto
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Koichiro Uchida
- Atopy Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kenta Fujio
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Ken Nagino
- Department of Hospital Administration, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Maria Miura
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Naoko Negishi
- Atopy Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Indoor Environment Neurophysiology Research, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yuichi Okumura
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Strategic Operating Room Management and Improvement, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yasutsugu Akasaki
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kunihiko Hirosawa
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Mizu Kuwahara
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Atsuko Eguchi
- Department of Hospital Administration, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hurramhon Shokirova
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Ai Yanagawa
- Department of Digital Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Akie Midorikawa-Inomata
- Department of Hospital Administration, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Akira Murakami
- Department of Ophthalmology, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Ophthalmology, Juntendo University Faculty of Medicine, Tokyo, Japan
- Department of Hospital Administration, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
4
|
Li Y, Tunbridge HM, Britton GJ, Hill EV, Sinai P, Cirillo S, Thompson C, Fallah-Arani F, Dovedi SJ, Wraith DC, Wülfing C. A LAT-Based Signaling Complex in the Immunological Synapse as Determined with Live Cell Imaging Is Less Stable in T Cells with Regulatory Capability. Cells 2021; 10:418. [PMID: 33671236 PMCID: PMC7921939 DOI: 10.3390/cells10020418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/12/2021] [Accepted: 02/12/2021] [Indexed: 12/03/2022] Open
Abstract
Peripheral immune regulation is critical for the maintenance of self-tolerance. Here we have investigated signaling processes that distinguish T cells with regulatory capability from effector T cells. The murine Tg4 T cell receptor recognizes a peptide derived from the self-antigen myelin basic protein. T cells from Tg4 T cell receptor transgenic mice can be used to generate effector T cells and three types of T cells with regulatory capability, inducible regulatory T cells, T cells tolerized by repeated in vivo antigenic peptide exposure or T cells treated with the tolerogenic drug UCB9608 (a phosphatidylinositol 4 kinase IIIβ inhibitor). We comparatively studied signaling in all of these T cells by activating them with the same antigen presenting cells presenting the same myelin basic protein peptide. Supramolecular signaling structures, as efficiently detected by large-scale live cell imaging, are critical mediators of T cell activation. The formation of a supramolecular signaling complex anchored by the adaptor protein linker for activation of T cells (LAT) was consistently terminated more rapidly in Tg4 T cells with regulatory capability. Such termination could be partially reversed by blocking the inhibitory receptors CTLA-4 and PD-1. Our work suggests that attenuation of proximal signaling may favor regulatory over effector function in T cells.
Collapse
Affiliation(s)
- Yikui Li
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
| | - Helen M Tunbridge
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
| | - Graham J Britton
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Elaine V Hill
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
| | - Parisa Sinai
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
| | - Silvia Cirillo
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
| | | | | | - Simon J Dovedi
- R&D Oncology, AstraZeneca, Granta Park, Cambridge, CB21 6GH, UK
| | - David C Wraith
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, B15 2TT, UK
| | - Christoph Wülfing
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, BS8 1TD, UK
| |
Collapse
|
5
|
Pinheiro DF, Szenes-Nagy AB, Maurano MM, Lietzenmayer M, Klicznik MM, Holly R, Kirchmeier D, Kitzmueller S, Achatz-Straussberger G, Rosenblum MD, Thalhamer J, Abbas AK, Gratz IK. Cutting Edge: Tissue Antigen Expression Levels Fine-Tune T Cell Differentiation Decisions In Vivo. THE JOURNAL OF IMMUNOLOGY 2020; 205:2577-2582. [PMID: 33037141 DOI: 10.4049/jimmunol.1901094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 09/11/2020] [Indexed: 11/19/2022]
Abstract
Immune homeostasis in peripheral tissues is, to a large degree, maintained by the differentiation and action of regulatory T cells (Treg) specific for tissue Ags. Using a novel mouse model, we have studied the differentiation of naive CD4+ T cells into Foxp3+ Treg in response to a cutaneous Ag (OVA). We found that expression of OVA resulted in fatal autoimmunity and in prevention of peripheral Treg generation. Inhibiting mTOR activity with rapamycin rescued the generation of Foxp3+ T cells. When we varied the level of Ag expression to modulate TCR signaling, we found that low Ag concentrations promoted the generation of Foxp3+ T cells, whereas high levels expanded effector T cells and caused severe autoimmunity. Our findings indicate that the expression level of tissue Ag is a key determinant of the balance between tissue-reactive effector and peripheral Foxp3+ T cells, which determines the choice between tolerance and autoimmunity.
Collapse
Affiliation(s)
- Douglas F Pinheiro
- Department of Biosciences, University of Salzburg, 5020 Salzburg, Austria
| | | | - Megan M Maurano
- Department of Biosciences, University of Salzburg, 5020 Salzburg, Austria.,Department of Pathology, University of California, San Francisco, San Francisco, CA 94143
| | | | - Maria M Klicznik
- Department of Biosciences, University of Salzburg, 5020 Salzburg, Austria
| | - Raimund Holly
- Department of Biosciences, University of Salzburg, 5020 Salzburg, Austria
| | - Daniel Kirchmeier
- Department of Biosciences, University of Salzburg, 5020 Salzburg, Austria
| | - Sophie Kitzmueller
- Department of Biosciences, University of Salzburg, 5020 Salzburg, Austria.,EB House Austria, Department of Dermatology, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria
| | | | - Michael D Rosenblum
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143; and
| | - Josef Thalhamer
- Department of Biosciences, University of Salzburg, 5020 Salzburg, Austria
| | - Abul K Abbas
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143
| | - Iris K Gratz
- Department of Biosciences, University of Salzburg, 5020 Salzburg, Austria; .,EB House Austria, Department of Dermatology, University Hospital of the Paracelsus Medical University, 5020 Salzburg, Austria.,Benaroya Research Institute, Seattle, WA 98101
| |
Collapse
|
6
|
Stanway JA, Isaacs JD. Tolerance-inducing medicines in autoimmunity: rheumatology and beyond. THE LANCET. RHEUMATOLOGY 2020; 2:e565-e575. [PMID: 38273619 DOI: 10.1016/s2665-9913(20)30100-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 04/09/2020] [Accepted: 04/09/2020] [Indexed: 12/14/2022]
Abstract
Autoimmunity is currently managed with generalised immunosuppression, which is associated with serious side-effects such as infection and cancer. An ideal treatment strategy would be to induce immune tolerance-ie, to reprogramme the immune system to stop recognising the host itself as a threat. Drug-free remission should follow such an intervention, representing a change in the approach to the treatment of autoimmune disease. Tolerance induction is achievable in animal models of autoimmunity but translation to the clinic has been slow. Nonetheless, progress has been made-eg, restoration of therapeutic responsiveness and drug-free remission have been achieved with stem cell transplantation in refractory autoimmunity, and significant delays in onset of type 1 diabetes in individuals at high risk have been achieved following a brief treatment with anti-CD3 monoclonal antibody. In the future, antigen-specific interventions should provide highly targeted, personalised approaches, avoiding generalised immunosuppression entirely. Such trials have already started, using both direct autoantigenic peptide administration, cellular therapies, and other modalities. In this Series paper, we discuss the history of immune tolerance induction with a focus on rheumatological disease while also highlighting essential data from other specialties. We propose key unanswered questions, which will be covered in other papers in this Series.
Collapse
Affiliation(s)
- James A Stanway
- National Institute for Health Research, Northern Deanery, Newcastle upon Tyne, UK; Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - John D Isaacs
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK; Musculoskeletal Unit, Newcastle upon Tyne Hospitals National Health Service Foundation Trust, Newcastle upon Tyne, UK.
| |
Collapse
|
7
|
Harper IG, Gjorgjimajkoska O, Siu JHY, Parmar J, Mulder A, Claas FHJ, Hosgood SA, Nicholson ML, Motallebzadeh R, Pettigrew GJ. Prolongation of allograft survival by passenger donor regulatory T cells. Am J Transplant 2019; 19:1371-1379. [PMID: 30548563 PMCID: PMC6519070 DOI: 10.1111/ajt.15212] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 11/13/2018] [Accepted: 11/17/2018] [Indexed: 01/25/2023]
Abstract
Tissue resident lymphocytes are present within many organs, and are presumably transferred at transplantation, but their impact on host immunity is unclear. Here, we examine whether transferred donor natural regulatory CD4 T cells (nT-regs) inhibit host alloimmunity and prolong allograft survival. Transfer of donor-strain lymphocytes was first assessed by identifying circulating donor-derived CD4 T cells in 21 consecutive human lung transplant recipients, with 3 patterns of chimerism apparent: transient, intermediate, and persistent (detectable for up to 6 weeks, 6 months, and beyond 1 year, respectively). The potential for transfer of donor nT-regs was then confirmed by analysis of leukocyte filters recovered from ex vivo normothermic perfusion circuits of human kidneys retrieved for transplantation. Finally, in a murine model of cardiac allograft vasculopathy, depletion of donor CD4 nT-regs before organ recovery resulted in markedly accelerated heart allograft rejection and augmented host effector antibody responses. Conversely, adoptive transfer or purified donor-strain nT-regs inhibited host humoral immunity and prolonged allograft survival, and more effectively so than following administration of recipient nT-regs. In summary, following transplantation, passenger donor-strain nT-regs can inhibit host adaptive immune responses and prolong allograft survival. Isolated donor-derived nT-regs may hold potential as a cellular therapy to improve transplant outcomes.
Collapse
Affiliation(s)
- Ines G. Harper
- Department of SurgerySchool of Clinical MedicineUniversity of CambridgeCambridgeUK
| | | | - Jacqueline H. Y. Siu
- Department of SurgerySchool of Clinical MedicineUniversity of CambridgeCambridgeUK
| | - Jasvir Parmar
- Department of Cardiothoracic TransplantationPapworth HospitalCambridgeUK
| | - Arend Mulder
- Department of Immunohaematology and Blood TransfusionLeiden University Medical CenterLeidenThe Netherlands
| | - Frans H. J. Claas
- Department of Immunohaematology and Blood TransfusionLeiden University Medical CenterLeidenThe Netherlands
| | - Sarah A. Hosgood
- Department of SurgerySchool of Clinical MedicineUniversity of CambridgeCambridgeUK
| | - Michael L. Nicholson
- Department of SurgerySchool of Clinical MedicineUniversity of CambridgeCambridgeUK
| | - Reza Motallebzadeh
- Centre for Surgical Innovation, Organ Repair & TransplantationUniversity College LondonLondonUK
- Centre for Transplantation, Department of Renal MedicineUniversity College LondonLondonUK
- Institute of Immunity and TransplantationUniversity College LondonLondonUK
| | - Gavin J. Pettigrew
- Department of SurgerySchool of Clinical MedicineUniversity of CambridgeCambridgeUK
| |
Collapse
|
8
|
Int"Dll"igent control of T-cell pathology in GVHD. Blood 2018; 132:2112-2114. [PMID: 30442748 DOI: 10.1182/blood-2018-09-875120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
9
|
Can the Fact That Myelin Proteins Are Old and Break down Explain the Origin of Multiple Sclerosis in Some People? J Clin Med 2018; 7:jcm7090281. [PMID: 30223497 PMCID: PMC6162792 DOI: 10.3390/jcm7090281] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 09/11/2018] [Accepted: 09/12/2018] [Indexed: 01/01/2023] Open
Abstract
Recent discoveries may change the way that multiple sclerosis (MS) is viewed, particularly with regard to the reasons for the untoward immune response. The fact that myelin proteins are long-lived, and that by the time we are adults, they are extensively degraded, alters our perspective on the reasons for the onset of autoimmunity and the origin of MS. For example, myelin basic protein (MBP) from every human brain past the age of 20 years, is so greatly modified, that it is effectively a different protein from the one that was laid down in childhood. Since only a subset of people with such degraded MBP develop MS, a focus on understanding the mechanism of immune responses to central nervous system (CNS) antigens and cerebral immune tolerance appear to be worthwhile avenues to explore. In accord with this, it will be productive to examine why all people, whose brains contain large quantities of a "foreign antigen", do not develop MS. Importantly for the potential causation of MS, MBP from MS patients breaks down differently from the MBP in aged controls. If the novel structures formed in these MS-specific regions are particularly antigenic, it could help explain the origin of MS. If verified, these findings could provide an avenue for the rational synthesis of drugs to prevent and treat MS.
Collapse
|
10
|
Neunkirchner A, Kratzer B, Köhler C, Smole U, Mager LF, Schmetterer KG, Trapin D, Leb-Reichl V, Rosloniec E, Naumann R, Kenner L, Jahn-Schmid B, Bohle B, Valenta R, Pickl WF. Genetic restriction of antigen-presentation dictates allergic sensitization and disease in humanized mice. EBioMedicine 2018; 31:66-78. [PMID: 29678672 PMCID: PMC6014064 DOI: 10.1016/j.ebiom.2018.04.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 03/29/2018] [Accepted: 04/02/2018] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Immunoglobulin(Ig)E-associated allergies result from misguided immune responses against innocuous antigens. CD4+ T lymphocytes are critical for initiating and perpetuating that process, yet the crucial factors determining whether an individual becomes sensitized towards a given allergen remain largely unknown. OBJECTIVE To determine the key factors for sensitization and allergy towards a given allergen. METHODS We here created a novel human T cell receptor(TCR) and human leucocyte antigen (HLA)-DR1 (TCR-DR1) transgenic mouse model of asthma, based on the human-relevant major mugwort (Artemisia vulgaris) pollen allergen Art v 1 to examine the critical factors for sensitization and allergy upon natural allergen exposure via the airways in the absence of systemic priming and adjuvants. RESULTS Acute allergen exposure led to IgE-independent airway hyperreactivity (AHR) and T helper(Th)2-prone lung inflammation in TCR-DR1, but not DR1, TCR or wildtype (WT) control mice, that was alleviated by prophylactic interleukin(IL)-2-αIL-2 mAb complex-induced expansion of Tregs. Chronic allergen exposure sensitized one third of single DR1 transgenic mice, however, without impacting on lung function. Similar treatment led to AHR and Th2-driven lung pathology in >90% of TCR-DR1 mice. Prophylactic and therapeutic expansion of Tregs with IL-2-αIL-2 mAb complexes blocked the generation and boosting of allergen-specific IgE associated with chronic allergen exposure. CONCLUSIONS We identify genetic restriction of allergen presentation as primary factor dictating allergic sensitization and disease against the major pollen allergen from the weed mugwort, which frequently causes sensitization and disease in humans. Furthermore, we demonstrate the importance of the balance between allergen-specific T effector and Treg cells for modulating allergic immune responses.
Collapse
Affiliation(s)
- Alina Neunkirchner
- Christian Doppler Laboratory for Immunomodulation, 1090 Vienna, Austria; Institute of Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Bernhard Kratzer
- Christian Doppler Laboratory for Immunomodulation, 1090 Vienna, Austria; Institute of Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Cordula Köhler
- Christian Doppler Laboratory for Immunomodulation, 1090 Vienna, Austria; Institute of Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Ursula Smole
- Institute of Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Lukas F Mager
- Institute of Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Klaus G Schmetterer
- Institute of Immunology, Medical University of Vienna, 1090 Vienna, Austria; Department of Laboratory Medicine, Medical University of Vienna, 1090 Vienna, Austria
| | - Doris Trapin
- Institute of Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | | | - Edward Rosloniec
- Department of Medicine, University of Tennessee Health Science Center, Memphis, 38163, TN, USA; Memphis Veterans Affairs Medical Center, 38104, TN, USA; Department of Pathology, University of Tennessee Health Science Center, Memphis, 38163, TN, USA
| | - Ronald Naumann
- Max Planck Institute for Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Lukas Kenner
- Department of Laboratory Animal Pathology, Medical University of Vienna, 1090 Vienna, Austria; Department of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria; Ludwig Boltzmann Institute for Cancer Research, Vienna, Austria
| | - Beatrice Jahn-Schmid
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, 1090 Vienna, Austria
| | - Barbara Bohle
- Christian Doppler Laboratory for Immunomodulation, 1090 Vienna, Austria; Department of Pathophysiology and Allergy Research, Medical University of Vienna, 1090 Vienna, Austria
| | - Rudolf Valenta
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, 1090 Vienna, Austria
| | - Winfried F Pickl
- Christian Doppler Laboratory for Immunomodulation, 1090 Vienna, Austria; Institute of Immunology, Medical University of Vienna, 1090 Vienna, Austria.
| |
Collapse
|
11
|
Gunasekera D, Zimring JC, Pratt KP. A unique major histocompatibility complex Class II-binding register correlates with HLA-DR11-associated immunogenicity of the major K blood group antigen. Transfusion 2018; 58:1171-1181. [PMID: 29464723 DOI: 10.1111/trf.14525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 11/29/2017] [Accepted: 01/02/2018] [Indexed: 11/27/2022]
Abstract
BACKGROUND Kell is a glycoprotein expressed on red blood cells (RBCs). Its K and k variants contain either Met (K antigen) or Thr (k antigen) at Position 193, respectively. Development of anti-K after K-mismatched antigen exposure via blood transfusions or pregnancy can destroy RBCs, leading to hemolytic transfusion reactions and hemolytic disease of the fetus and newborn. The immunogenicity of overlapping 15-mer Kell peptides with M193 or T193 at every possible position was investigated previously. Interestingly, Peptide W179 to M193, with the polymorphic M193T residue at the peptide's C-terminus, was the most effective at stimulating CD4 T cells from a series of K-immunized women. STUDY DESIGN AND METHODS This study investigates the basis for HLA restriction of anti-K immune responses. Major histocompatibility complex Class II (MHCII)-binding prediction algorithms and quantitative peptide-MHCII-binding assays were employed to determine the binding registers; anchor residues; and affinities of wild-type, truncated, and sequence-modified K and k peptides. Predictions were generated using Immune Epitope Database and ProPred algorithms. Competitive peptide-MHCII-binding assays utilized 12 recombinant HLA-DR proteins, K and k peptides, and high-affinity MHCII-restricted reference peptides. RESULTS The peptide-MHCII-binding assays identified a unique K peptide-binding register (W179-S187) restricted to HLA-DRB1*11:01, in addition to partially overlapping binding registers that included the K/k M193T polymorphic site and that bound promiscuously to multiple HLA-DR proteins. CONCLUSION Three partially overlapping MHCII-binding motifs for HLA-DRB1*11:01 result in high-avidity K-peptide binding, which may contribute to HLA-DR11-restricted immunogenicity associated with the K allele.
Collapse
Affiliation(s)
- Devi Gunasekera
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| | | | - Kathleen P Pratt
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland
| |
Collapse
|
12
|
Emmanouil M, Tseveleki V, Triantafyllakou I, Nteli A, Tselios T, Probert L. A Cyclic Altered Peptide Analogue Based on Myelin Basic Protein 87-99 Provides Lasting Prophylactic and Therapeutic Protection Against Acute Experimental Autoimmune Encephalomyelitis. Molecules 2018; 23:molecules23020304. [PMID: 29385090 PMCID: PMC6017753 DOI: 10.3390/molecules23020304] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 01/18/2018] [Accepted: 01/24/2018] [Indexed: 12/24/2022] Open
Abstract
In this report, amide-linked cyclic peptide analogues of the 87-99 myelin basic protein (MBP) epitope, a candidate autoantigen in multiple sclerosis (MS), are tested for therapeutic efficacy in experimental autoimmune encephalomyelitis (EAE). Cyclic altered peptide analogues of MBP87-99 with substitutions at positions 91 and/or 96 were tested for protective effects when administered using prophylactic or early therapeutic protocols in MBP72-85-induced EAE in Lewis rats. The Lys91 and Pro96 of MBP87-99 are crucial T-cell receptor (TCR) anchors and participate in the formation of trimolecular complex between the TCR-antigen (peptide)-MHC (major histocompability complex) for the stimulation of encephalitogenic T cells that are necessary for EAE induction and are implicated in MS. The cyclic peptides were synthesized using Solid Phase Peptide Synthesis (SPPS) applied on the 9-fluorenylmethyloxycarboxyl/tert-butyl Fmoc/tBu methodology and combined with the 2-chlorotrityl chloride resin (CLTR-Cl). Cyclo(91-99)[Ala96]MBP87-99, cyclo(87-99)[Ala91,96]MBP87-99 and cyclo(87-99)[Arg91, Ala96]MBP87-99, but not wild-type linear MBP87-99, strongly inhibited MBP72-85-induced EAE in Lewis rats when administered using prophylactic and early therapeutic vaccination protocols. In particular, cyclo(87-99)[Arg91, Ala96]MBP87-99 was highly effective in preventing the onset and development of clinical symptoms and spinal cord pathology and providing lasting protection against EAE induction.
Collapse
MESH Headings
- Animals
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Female
- Myelin Basic Protein/chemical synthesis
- Myelin Basic Protein/chemistry
- Myelin Basic Protein/pharmacology
- Peptide Fragments/chemical synthesis
- Peptide Fragments/chemistry
- Peptide Fragments/pharmacology
- Peptides, Cyclic/chemical synthesis
- Peptides, Cyclic/chemistry
- Peptides, Cyclic/pharmacology
- Rats
- Rats, Inbred Lew
Collapse
Affiliation(s)
- Mary Emmanouil
- Laboratory of Molecular Genetics, Hellenic Pasteur Institute, 127 Vasilissis Sophias Ave., 11521 Athens, Greece.
| | - Vivian Tseveleki
- Laboratory of Molecular Genetics, Hellenic Pasteur Institute, 127 Vasilissis Sophias Ave., 11521 Athens, Greece.
| | | | - Agathi Nteli
- Department of Chemistry, University of Patras, 26504 Patras, Greece.
| | - Theodore Tselios
- Department of Chemistry, University of Patras, 26504 Patras, Greece.
| | - Lesley Probert
- Laboratory of Molecular Genetics, Hellenic Pasteur Institute, 127 Vasilissis Sophias Ave., 11521 Athens, Greece.
| |
Collapse
|
13
|
Chirumbolo S, Bjørklund G, Sboarina A, Vella A. The role of basophils as innate immune regulatory cells in allergy and immunotherapy. Hum Vaccin Immunother 2018; 14:815-831. [PMID: 29257936 DOI: 10.1080/21645515.2017.1417711] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Basophils are circulating cells that are associated quite exclusively with allergy response and hypersensitivity reactions but their role in the immune network might be much more intriguing and complex than previously expected. The feasibility of testing their biology in vitro for allergy research and diagnosis, due fundamentally to their quite easy availability in the peripheral blood, made them the major source for assessing allergy in the laboratory assay, when yet many further cells such as mast cells and eosinophils are much more involved as effector cells in allergy than circulating basophils. Interestingly, basophil numbers change rarely in peripheral blood during an atopic response, while we might yet observe an increase in eosinophils and modification in the biology of mast cells in the tissue during an hypersensitivity response. Furthermore, the fact that basophils are very scanty in numbers suggests that they should mainly serve as regulatory cells in immunity, rather than effector leukocytes, as still believed by the majority of physicians. In this review we will try to describe and elucidate the possible role of these cells, known as "innate IL4-producing cells" in the immune regulation of allergy and their function in allergen immunotherapy.
Collapse
Affiliation(s)
- Salvatore Chirumbolo
- a Department of Neurological and Movement Sciences , University of Verona , Verona , Italy
| | - Geir Bjørklund
- b Council for Nutritional and Environmental Medicine (CONEM) , Mo i Rana , Norway
| | - Andrea Sboarina
- c Department of Surgery , Dentistry, Paediatrics and Gynaecology-University of Verona , Verona , Italy
| | - Antonio Vella
- d Unit of Immunology-Azienda Ospedaliera Universitaria Integrata (AOUI) , Verona , Italy
| |
Collapse
|
14
|
A humanized HLA-DR4 mouse model for autoimmune myocarditis. J Mol Cell Cardiol 2017; 107:22-26. [PMID: 28431892 PMCID: PMC5466360 DOI: 10.1016/j.yjmcc.2017.04.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 04/05/2017] [Accepted: 04/17/2017] [Indexed: 11/20/2022]
Abstract
Myocarditis, the principal cause of dilated cardiomyopathy and heart failure in young adults, is associated with autoimmunity to human cardiac α-myosin (hCAM) and the DR4 allele of human major histocompatibility II (MHCII). We developed an hCAM-induced myocarditis model in human HLA-DR4 transgenic mice that lack all mouse MHCII genes, demonstrating that immunization for 3 weeks significantly increased splenic T-cell proliferative responses and titres of IgG1 and IgG2c antibodies, abolished weight gain, provoked cardiac inflammation and significantly impaired cardiac output and fractional shortening, by echocardiography, compared to adjuvant-injected mice. Neither cardiac dilatation nor fibrosis occurred at this time point but prolonging the experiment was associated with mortality. Treatment with mixtures of hCAM derived peptides predicted to have high affinity for DR4 significantly preserved ejection fraction and fractional shortening. Our new humanized mouse model of autoimmune cardiomyopathy should be useful to refine hCAM-derived peptide treatment. We developed a novel fully-humanized mouse model of autoimmune myocarditis. Human leukocyte antigen DR4 transgenic mice were immunized with human cardiac myosin. After 3 weeks, mice developed myocarditis and impaired myocardial function. A pilot study established the feasibility of peptide immunotherapy in this model.
Collapse
|
15
|
da Silva MB, da Cunha FF, Terra FF, Camara NOS. Old game, new players: Linking classical theories to new trends in transplant immunology. World J Transplant 2017; 7:1-25. [PMID: 28280691 PMCID: PMC5324024 DOI: 10.5500/wjt.v7.i1.1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 11/16/2016] [Accepted: 12/07/2016] [Indexed: 02/05/2023] Open
Abstract
The evolutionary emergence of an efficient immune system has a fundamental role in our survival against pathogenic attacks. Nevertheless, this same protective mechanism may also establish a negative consequence in the setting of disorders such as autoimmunity and transplant rejection. In light of the latter, although research has long uncovered main concepts of allogeneic recognition, immune rejection is still the main obstacle to long-term graft survival. Therefore, in order to define effective therapies that prolong graft viability, it is essential that we understand the underlying mediators and mechanisms that participate in transplant rejection. This multifaceted process is characterized by diverse cellular and humoral participants with innate and adaptive functions that can determine the type of rejection or promote graft acceptance. Although a number of mediators of graft recognition have been described in traditional immunology, recent studies indicate that defining rigid roles for certain immune cells and factors may be more complicated than originally conceived. Current research has also targeted specific cells and drugs that regulate immune activation and induce tolerance. This review will give a broad view of the most recent understanding of the allogeneic inflammatory/tolerogenic response and current insights into cellular and drug therapies that modulate immune activation that may prove to be useful in the induction of tolerance in the clinical setting.
Collapse
|
16
|
Mo LH, Yang LT, Zeng L, Xu LZ, Zhang HP, Li LJ, Liu JQ, Xiao XJ, Zheng PY, Liu ZG, Yang PC. Dust mite allergen, glutathione S-transferase, induces T cell immunoglobulin mucin domain-4 in dendritic cells to facilitate initiation of airway allergy. Clin Exp Allergy 2016; 47:264-270. [PMID: 27532130 DOI: 10.1111/cea.12800] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Revised: 07/03/2016] [Accepted: 07/17/2016] [Indexed: 12/19/2022]
Abstract
BACKGROUND Allergens from dust mites play a critical role in the pathogenesis of airway allergy. The mechanism by which dust mite allergens induce allergic diseases is not fully understood yet. OBJECTIVE This study tests a hypothesis that the eighth subtypes of Dermatophagoides farina allergen (Derf8) play an important role in the induction of airway allergy. METHODS The protein of Derf8 was synthesized via molecular cloning approach. Dendritic cells (DC) were stimulated with Derf8 in the culture, and then, the expression of T cell immunoglobulin mucin domain 4 (TIM4) in dendritic cells (DC) was analysed. The role of Derf8 in the induction of airway allergy was evaluated with a mouse model. RESULTS Exposure to Derf8 markedly induced the TIM4 expression in DCs by modulating the chromatin at the TIM4 promoter locus. Derf8 played a critical role in the expansion of the T helper 2 response in the mouse airway via inducing DCs to produce TIM4. Administration with Derf8-depleted dust mite extracts (DME) inhibited the allergic inflammation and induced regulatory T cells in mice with airway allergy. CONCLUSION Derf8 plays an important role in the initiation of dust mite allergy. Vaccination with Derf8-deficient DME is more efficient to inhibit the dust mite allergic inflammation than using wild DME.
Collapse
Affiliation(s)
- L-H Mo
- The Research Center of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen, China.,Longgang ENT Hospital, Shenzhen, China
| | - L-T Yang
- The Research Center of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen, China.,Longgang ENT Hospital, Shenzhen, China.,Brain Body Institute, McMaster University, Hamilton, ON, Canada
| | - L Zeng
- The Research Center of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - L-Z Xu
- The Research Center of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - H-P Zhang
- Brain Body Institute, McMaster University, Hamilton, ON, Canada
| | - L-J Li
- Brain Body Institute, McMaster University, Hamilton, ON, Canada
| | - J-Q Liu
- The Research Center of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen, China.,Longgang ENT Hospital, Shenzhen, China.,Brain Body Institute, McMaster University, Hamilton, ON, Canada
| | - X-J Xiao
- The Research Center of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - P-Y Zheng
- Department of Gastroenterology, The Fifth Hospital, Zhengzhou University, Zhengzhou, China
| | - Z-G Liu
- The Research Center of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - P-C Yang
- The Research Center of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen, China
| |
Collapse
|
17
|
Poly-N-Acetylglucosamine Production by Staphylococcus epidermidis Cells Increases Their In Vivo Proinflammatory Effect. Infect Immun 2016; 84:2933-43. [PMID: 27481237 DOI: 10.1128/iai.00290-16] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 07/21/2016] [Indexed: 12/12/2022] Open
Abstract
Poly-N-acetylglucosamine (PNAG) is a major component of the Staphylococcus epidermidis biofilm extracellular matrix. However, it is not yet clear how this polysaccharide impacts the host immune response and infection-associated pathology. Faster neutrophil recruitment and bacterial clearance were observed in mice challenged intraperitoneally with S. epidermidis biofilm cells of the PNAG-producing 9142 strain than in mice similarly challenged with the isogenic PNAG-defective M10 mutant. Moreover, intraperitoneal priming with 9142 cells exacerbated liver inflammatory pathology induced by a subsequent intravenous S. epidermidis challenge, compared to priming with M10 cells. The 9142-primed mice had elevated splenic CD4(+) T cells producing gamma interferon and interleukin-17A, indicating that PNAG promoted cell-mediated immunity. Curiously, despite having more marked liver tissue pathology, 9142-primed mice also had splenic T regulatory cells with greater suppressive activity than those of their M10-primed counterparts. By showing that PNAG production by S. epidermidis biofilm cells exacerbates host inflammatory pathology, these results together suggest that this polysaccharide contributes to the clinical features associated with biofilm-derived infections.
Collapse
|
18
|
Zhai T, Sun Y, Li H, Zhang J, Huo R, Li H, Shen B, Li N. Unique immunomodulatory effect of paeoniflorin on type I and II macrophages activities. J Pharmacol Sci 2016; 130:143-50. [PMID: 26852260 DOI: 10.1016/j.jphs.2015.12.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 12/19/2015] [Accepted: 12/25/2015] [Indexed: 10/22/2022] Open
Abstract
It has been widely accepted that macrophages are divided into M1 "pro-inflammatory" macrophages and M2 "anti-inflammatory" macrophages and an uncontrolled macrophage polarization plays an important role in the pathogenesis of different diseases. As the main substance of total glucosides of peony, paeoniflorin (PF), has been widely used to treat autoimmune and autoinflammatory diseases for years. Mechanistically, PF has been found to alter activities of many immune cells, which could further reduce inflammation and tissue damage. However, whether and how PF affects macrophages activities in vitro remains unknown. In current study, using M1 and M2 cells generated from mouse bone marrow precursors, we explored the role of PF in regulating M1/M2 cells activity in vitro. The results showed that PF inhibited LPS-induced M1 activity by reducing iNOS expression and NO production via decreasing LPS/NF-κB signaling pathway; whereas, PF enhanced IL-4-provoked M2 function by up-regulating Arg-1 production and activity via increasing IL-4/STAT6 signaling pathway. Our new finding indicates that PF can suppress M1 cells activity and enhance M2 cells function simultaneously, which could help to ameliorate autoimmune and autoinflammatory diseases in clinical treatment.
Collapse
Affiliation(s)
- Tianhang Zhai
- Shanghai Institute of Immunology & Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yue Sun
- Shanghai Institute of Immunology & Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Department of Rheumatology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Huidan Li
- Shanghai Institute of Immunology & Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Zhang
- Shanghai Institute of Immunology & Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rongfen Huo
- Shanghai Institute of Immunology & Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haichuan Li
- Shanghai Institute of Immunology & Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Maternity and Child Health Hospital, Changning, Shanghai, China
| | - Baihua Shen
- Shanghai Institute of Immunology & Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ningli Li
- Shanghai Institute of Immunology & Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
19
|
Swann JW, Garden OA. Novel immunotherapies for immune-mediated haemolytic anaemia in dogs and people. Vet J 2016; 207:13-9. [DOI: 10.1016/j.tvjl.2015.10.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
20
|
Zhang D, Tu E, Kasagi S, Zanvit P, Chen Q, Chen W. Manipulating regulatory T cells: a promising strategy to treat autoimmunity. Immunotherapy 2015; 7:1201-11. [PMID: 26568117 DOI: 10.2217/imt.15.79] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
CD4(+)CD25(+)Foxp3(+)regulatory T cells (Treg cells) are extremely important in maintaining immune tolerance. Manipulation of Treg cells, especially autoantigen-specific Treg cells is a promising approach for treatments of autoimmune disease since Treg cells may provide the advantage of antigen specificity without overall immune suppression. However, the clinical application of Treg cells has long been limited due to low numbers of Treg cells and the difficulty in identifying their antigen specificity. In this review, we summarize studies that demonstrate regression of autoimmune diseases using Treg cells as therapeutics. We also discuss approaches to generate polyclonal and autoantigen-specific Treg cells in vitro and in vivo. We also discuss our recent study that describes a novel approach of generating autoantigen-specific Treg cells in vivo and restoring immune tolerance by two steps apoptosis-antigen therapy.
Collapse
Affiliation(s)
- Dunfang Zhang
- Mucosal Immunology Section, OPCB, National Institute of Dental & Craniofacial Research, NIH, Bethesda, MD 20892, USA.,State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Eric Tu
- Mucosal Immunology Section, OPCB, National Institute of Dental & Craniofacial Research, NIH, Bethesda, MD 20892, USA
| | - Shimpei Kasagi
- Mucosal Immunology Section, OPCB, National Institute of Dental & Craniofacial Research, NIH, Bethesda, MD 20892, USA
| | - Peter Zanvit
- Mucosal Immunology Section, OPCB, National Institute of Dental & Craniofacial Research, NIH, Bethesda, MD 20892, USA
| | - Qianming Chen
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - WanJun Chen
- Mucosal Immunology Section, OPCB, National Institute of Dental & Craniofacial Research, NIH, Bethesda, MD 20892, USA
| |
Collapse
|
21
|
Fryer M, Grahammer J, Khalifian S, Furtmüller GJ, Lee WPA, Raimondi G, Brandacher G. Exploring cell-based tolerance strategies for hand and face transplantation. Expert Rev Clin Immunol 2015; 11:1189-204. [DOI: 10.1586/1744666x.2015.1078729] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|