1
|
Langguth P, Peckert-Maier K, Beck P, Kuhnt C, Draßner C, Deinzer A, Steinkasserer A, Wild AB. CD83 acts as immediate early response gene in activated macrophages and exhibits specific intracellular trafficking properties. Biochem Biophys Res Commun 2023; 647:37-46. [PMID: 36709671 DOI: 10.1016/j.bbrc.2023.01.069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 01/20/2023] [Indexed: 01/24/2023]
Abstract
Macrophages (MΦ) are remarkably plastic cells, which assume phenotypes in every shade between a pro-inflammatory classical activation, and anti-inflammatory or resolving activation. Therefore, elucidation of mechanisms involved in shaping MΦ plasticity and function is key to understand their role during immunological balance. The immune-modulating CD83 molecule is expressed on activated immune cells and various tissue resident MΦ, rendering it an interesting candidate for affecting MΦ biology. However, in-depth analyses of the precise kinetics and trafficking of CD83 within pro-inflammatory, LPS activated bone-marrow-derived MΦ have not been performed. In this study, we show that activation with LPS leads to a very fast and strong, but transient increase of CD83 expression on these cells. Its expression peaks within 2 h of stimulation and is thereby faster than the early activation antigen CD69. To trace the CD83 trafficking through MΦs, we employed multiple inhibitors, thereby revealing a de novo synthesis and transport of the protein to the cell surface followed by lysosomal degradation, all within 6 h. Moreover, we found a similar expression kinetic and trafficking in human monocyte derived MΦ. This places CD83 at a very early point of MΦ activation suggesting an important role in decisions regarding the subsequent cellular fate.
Collapse
Affiliation(s)
- Pia Langguth
- Department of Immune Modulation, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| | - Katrin Peckert-Maier
- Department of Immune Modulation, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Philipp Beck
- Department of Immune Modulation, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Christine Kuhnt
- Department of Immune Modulation, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Christina Draßner
- Department of Immune Modulation, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Andrea Deinzer
- Institute of Microbiology - Clinical Microbiology, Immunology and Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander -Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Alexander Steinkasserer
- Department of Immune Modulation, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Andreas B Wild
- Department of Immune Modulation, Universitätsklinikum Erlangen and Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
2
|
Royzman D, Peckert-Maier K, Stich L, König C, Wild AB, Tauchi M, Ostalecki C, Kiesewetter F, Seyferth S, Lee G, Eming SA, Fuchs M, Kunz M, Stürmer EK, Peters EMJ, Berking C, Zinser E, Steinkasserer A. Soluble CD83 improves and accelerates wound healing by the induction of pro-resolving macrophages. Front Immunol 2022; 13:1012647. [PMID: 36248909 PMCID: PMC9564224 DOI: 10.3389/fimmu.2022.1012647] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 09/13/2022] [Indexed: 11/22/2022] Open
Abstract
To facilitate the recovery process of chronic and hard-to-heal wounds novel pro-resolving treatment options are urgently needed. We investigated the pro-regenerative properties of soluble CD83 (sCD83) on cutaneous wound healing, where sCD83 accelerated wound healing not only after systemic but also after topical application, which is of high therapeutic interest. Cytokine profile analyses revealed an initial upregulation of inflammatory mediators such as TNFα and IL-1β, followed by a switch towards pro-resolving factors, including YM-1 and IL-10, both expressed by tissue repair macrophages. These cells are known to mediate resolution of inflammation and stimulate wound healing processes by secretion of growth factors such as epidermal growth factor (EGF) and vascular endothelial growth factor (VEGF), which promote vascularization as well as fibroblast and keratinocyte differentiation. In conclusion, we have found strong wound healing capacities of sCD83 beyond the previously described role in transplantation and autoimmunity. This makes sCD83 a promising candidate for the treatment of chronic- and hard-to-heal wounds.
Collapse
Affiliation(s)
- Dmytro Royzman
- Department of Immune Modulation, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- *Correspondence: Dmytro Royzman, ; Alexander Steinkasserer,
| | - Katrin Peckert-Maier
- Department of Immune Modulation, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Lena Stich
- Department of Immune Modulation, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Christina König
- Department of Immune Modulation, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Andreas B. Wild
- Department of Immune Modulation, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Miyuki Tauchi
- Department of Internal Medicine 2, University Hospital Erlangen, FAU, Erlangen, Germany
| | - Christian Ostalecki
- Department of Dermatology, University Hospital Erlangen, FAU, Erlangen, Germany
| | | | - Stefan Seyferth
- Division of Pharmaceutics, Department of Chemistry and Pharmacy, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Geoffrey Lee
- Division of Pharmaceutics, Department of Chemistry and Pharmacy, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Sabine A. Eming
- Department of Dermatology, University Hospital Cologne, Center for Molecular Medicine Cologne (CMMC), Cologne Excellence Cluster Cluster of Excellence for Aging Research (CECAD), University of Cologne, Cologne, Germany
| | - Maximilian Fuchs
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hannover, Germany
| | - Meik Kunz
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hannover, Germany
- Department of Medical Informatics, Friedrich-Alexander University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Germany
| | - Ewa K. Stürmer
- Department for Vascular Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Eva M. J. Peters
- Psychoneuroimmunology Laboratory, Klinik für Psychosomatik und Psychotherapie, Justus-Liebig Universität Gießen, Gießen, Germany
| | - Carola Berking
- Department of Dermatology, University Hospital Erlangen, FAU, Erlangen, Germany
| | - Elisabeth Zinser
- Department of Immune Modulation, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Alexander Steinkasserer
- Department of Immune Modulation, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
- *Correspondence: Dmytro Royzman, ; Alexander Steinkasserer,
| |
Collapse
|
3
|
Peckert-Maier K, Schönberg A, Wild AB, Royzman D, Braun G, Stich L, Hadrian K, Tripal P, Cursiefen C, Steinkasserer A, Zinser E, Bock F. Pre-incubation of corneal donor tissue with sCD83 improves graft survival via the induction of alternatively activated macrophages and tolerogenic dendritic cells. Am J Transplant 2022; 22:438-454. [PMID: 34467638 DOI: 10.1111/ajt.16824] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 08/02/2021] [Accepted: 08/22/2021] [Indexed: 01/25/2023]
Abstract
Immune responses reflect a complex interplay of cellular and extracellular components which define the microenvironment of a tissue. Therefore, factors that locally influence the microenvironment and re-establish tolerance might be beneficial to mitigate immune-mediated reactions, including the rejection of a transplant. In this study, we demonstrate that pre-incubation of donor tissue with the immune modulator soluble CD83 (sCD83) significantly improves graft survival using a high-risk corneal transplantation model. The induction of tolerogenic mechanisms in graft recipients was achieved by a significant upregulation of Tgfb, Foxp3, Il27, and Il10 in the transplant and an increase of regulatory dendritic cells (DCs), macrophages (Mφ), and T cells (Tregs) in eye-draining lymph nodes. The presence of sCD83 during in vitro DC and Mφ generation directed these cells toward a tolerogenic phenotype leading to reduced proliferation-stimulating activity in MLRs. Mechanistically, sCD83 induced a tolerogenic Mφ and DC phenotype, which favors Treg induction and significantly increased transplant survival after adoptive cell transfer. Conclusively, pre-incubation of corneal grafts with sCD83 significantly prolongs graft survival by modulating recipient Mφ and DCs toward tolerance and thereby establishing a tolerogenic microenvironment. This functional strategy of donor graft pre-treatment paves the way for new therapeutic options in the field of transplantation.
Collapse
Affiliation(s)
- Katrin Peckert-Maier
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Alfrun Schönberg
- Department of Experimental Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Andreas B Wild
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Dmytro Royzman
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Gabriele Braun
- Department of Experimental Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Lena Stich
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Karina Hadrian
- Department of Experimental Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Philipp Tripal
- Optical Imaging Centre, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Claus Cursiefen
- Department of Experimental Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | | | - Elisabeth Zinser
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Felix Bock
- Department of Experimental Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
4
|
Peckert-Maier K, Royzman D, Langguth P, Marosan A, Strack A, Sadeghi Shermeh A, Steinkasserer A, Zinser E, Wild AB. Tilting the Balance: Therapeutic Prospects of CD83 as a Checkpoint Molecule Controlling Resolution of Inflammation. Int J Mol Sci 2022; 23:732. [PMID: 35054916 PMCID: PMC8775349 DOI: 10.3390/ijms23020732] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 01/05/2022] [Accepted: 01/08/2022] [Indexed: 12/15/2022] Open
Abstract
Chronic inflammatory diseases and transplant rejection represent major challenges for modern health care. Thus, identification of immune checkpoints that contribute to resolution of inflammation is key to developing novel therapeutic agents for those conditions. In recent years, the CD83 (cluster of differentiation 83) protein has emerged as an interesting potential candidate for such a "pro-resolution" therapy. This molecule occurs in a membrane-bound and a soluble isoform (mCD83 and sCD83, respectively), both of which are involved in resolution of inflammation. Originally described as a maturation marker on dendritic cells (DCs), mCD83 is also expressed by activated B and T cells as well as regulatory T cells (Tregs) and controls turnover of MHC II molecules in the thymus, and thereby positive selection of CD4+ T cells. Additionally, it serves to confine overshooting (auto-)immune responses. Consequently, animals with a conditional deletion of CD83 in DCs or regulatory T cells suffer from impaired resolution of inflammation. Pro-resolving effects of sCD83 became evident in pre-clinical autoimmune and transplantation models, where application of sCD83 reduced disease symptoms and enhanced allograft survival, respectively. Here, we summarize recent advances regarding CD83-mediated resolution of inflammatory responses, its binding partners as well as induced signaling pathways, and emphasize its therapeutic potential for future clinical trials.
Collapse
Affiliation(s)
- Katrin Peckert-Maier
- Department of Immune Modulation, Universitätsklinikum Erlangen, Friedrich-Alexander Universität—Erlangen-Nürnberg, 91052 Erlangen, Germany; (D.R.); (P.L.); (A.M.); (A.S.); (A.S.S.); (A.S.); (E.Z.)
| | | | | | | | | | | | | | | | - Andreas B. Wild
- Department of Immune Modulation, Universitätsklinikum Erlangen, Friedrich-Alexander Universität—Erlangen-Nürnberg, 91052 Erlangen, Germany; (D.R.); (P.L.); (A.M.); (A.S.); (A.S.S.); (A.S.); (E.Z.)
| |
Collapse
|
5
|
Xiong L, Wang D, Lin S, Wang Y, Luo M, Gao L. Soluble CD83 inhibits acute rejection by up regulating TGF-β and IDO secretion in rat liver transplantation. Transpl Immunol 2021; 64:101351. [PMID: 33171217 DOI: 10.1016/j.trim.2020.101351] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 10/31/2020] [Accepted: 10/31/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Allogeneic transplantation immune tolerance is currently a hot research issue and soluble CD83(sCD83) is a novel immunomodulator with great potential in inducing transplantation tolerance. OBJECTIVE To investigate the mechanism of the immune tolerance effect of sCD83 on rat liver transplantation. METHOD A rat liver transplantation model was established to study the effects of sCD83 on the expression levels of IL-2, IL-10, and TGF-β in peripheral blood and the mRNA expressions of foxp3, TGF-β, and Indoleamine 2,3-dioxygenase (IDO) in liver. The expression changes of costimulatory molecules CD80, CD86, and MHC-II on the surface of DC cells and the expressions of IDO + DC cell, TGF-β + CD4 + T cell, and CD4 + CD25 + Foxp3 + T cell were analyzed and compared. RESULTS sCD83 alleviated the rejection activity index (RAI) of rat liver transplantation in the early stage, increased the expressions of TGF-β, IL-10 in peripheral blood and the mRNAs of IDO, TGF-β and foxp3 in the transplanted liver, and down-regulated the expressions of MHC-II, CD86, and CD80 in DC cells, resulting in significant increased numbers of tolerogenic TGF-β + CD4 + T cells, Treg cells, and IDO + DC cells with low expression. CONCLUSION sCD83 inhibited acute rejection after liver transplantation in an allogeneic rat, and the mechanism was associated with the effect that sCD83 increased the expression of TGF-β, activated IDO immunosuppressive pathway, and increased tolerogenic DC cells and Treg cells.
Collapse
Affiliation(s)
- Liangxing Xiong
- The First Affiliated Hospital of Hainan Medical College, China; The Third Affiliated Hospital of Sun Yat-sen University Yuedong Hospital, China
| | - Danxin Wang
- The First Affiliated Hospital of Hainan Medical College, China
| | - Shibu Lin
- The First Affiliated Hospital of Hainan Medical College, China
| | - Yubin Wang
- The First Affiliated Hospital of Hainan Medical College, China
| | - Mingwei Luo
- The First Affiliated Hospital of Hainan Medical College, China
| | - Lianghui Gao
- The First Affiliated Hospital of Hainan Medical College, China.
| |
Collapse
|
6
|
Huo S, Wu F, Zhang J, Wang X, Li W, Cui D, Zuo Y, Hu M, Zhong F. Porcine soluble CD83 alleviates LPS-induced abortion in mice by promoting Th2 cytokine production, Treg cell generation and trophoblast invasion. Theriogenology 2020; 157:149-161. [PMID: 32810792 DOI: 10.1016/j.theriogenology.2020.07.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 07/16/2020] [Accepted: 07/25/2020] [Indexed: 01/24/2023]
Abstract
CD83, either in its membrance-bound form (mCD83) or soluble form (sCD83), is an important immunomodulatory molecule in humans and mice. While mCD83 is immunostimulatory, sCD83 exhibits striking immunosuppressive activities, suggesting that sCD83 may be used to combat inflammatory diseases, such as rheumatoid arthritis, graft-versus-host disease and habitual abortion. Although many studies had shed lights on the role of CD83 in humans and mice, little is known about CD83 in other animals. Recently, we showed that porcine CD83 had similar biochemical characteristics and immunoregulatory functions as its human counterpart. However, whether porcine sCD83 (psCD83) is involved in maintaining the immunological tolerance at the maternal-fetal interface and thereby prevents embryo loss and abortion during pregnancy is unclear. In this study, we used LPS-induced animal model to analyze the effect of porcine sCD83 on the mouse abortion. Results showed that psCD83 could significantly alleviate LPS-induced abortion in mice, indicating that the psCD83 had the function of fetal protection. Mechanically, psCD83-mediated fetal protection was related to the promotion on Th2 cytokine production, Treg cell differentiation and trophoblast invasion. This study provides a molecular basis for the fetal protection of psCD83, as well as a potential target for the regulation of maternal-fetal interfacial immune tolerance.
Collapse
Affiliation(s)
- Shanshan Huo
- College of Animal Science and Technology and College of Veterinary Medicine, Hebei Agricultural University, 289 Lingyusi Streat, Baoding, Hebei, 071000, China; Hebei Veterinary Biotechnology Innovation Center, Hebei Agricultural University, 289 Lingyusi Streat, Baoding, 071001, China.
| | - Fengyang Wu
- College of Animal Science and Technology and College of Veterinary Medicine, Hebei Agricultural University, 289 Lingyusi Streat, Baoding, Hebei, 071000, China.
| | - Jianlou Zhang
- College of Animal Science and Technology and College of Veterinary Medicine, Hebei Agricultural University, 289 Lingyusi Streat, Baoding, Hebei, 071000, China; Hebei Veterinary Biotechnology Innovation Center, Hebei Agricultural University, 289 Lingyusi Streat, Baoding, 071001, China.
| | - Xing Wang
- College of Animal Science and Technology and College of Veterinary Medicine, Hebei Agricultural University, 289 Lingyusi Streat, Baoding, Hebei, 071000, China.
| | - Wenyan Li
- College of Animal Science and Technology and College of Veterinary Medicine, Hebei Agricultural University, 289 Lingyusi Streat, Baoding, Hebei, 071000, China; Department of Biology, College of Basic Medicine, Hebei University, 180 Wusi Dong Road, Baoding, Hebei, 071000, China.
| | - Dan Cui
- College of Animal Science and Technology and College of Veterinary Medicine, Hebei Agricultural University, 289 Lingyusi Streat, Baoding, Hebei, 071000, China.
| | - Yuzhu Zuo
- College of Animal Science and Technology and College of Veterinary Medicine, Hebei Agricultural University, 289 Lingyusi Streat, Baoding, Hebei, 071000, China; Hebei Veterinary Biotechnology Innovation Center, Hebei Agricultural University, 289 Lingyusi Streat, Baoding, 071001, China.
| | - Man Hu
- College of Animal Science and Technology and College of Veterinary Medicine, Hebei Agricultural University, 289 Lingyusi Streat, Baoding, Hebei, 071000, China; Hebei Veterinary Biotechnology Innovation Center, Hebei Agricultural University, 289 Lingyusi Streat, Baoding, 071001, China.
| | - Fei Zhong
- College of Animal Science and Technology and College of Veterinary Medicine, Hebei Agricultural University, 289 Lingyusi Streat, Baoding, Hebei, 071000, China; Hebei Veterinary Biotechnology Innovation Center, Hebei Agricultural University, 289 Lingyusi Streat, Baoding, 071001, China.
| |
Collapse
|
7
|
Grosche L, Knippertz I, König C, Royzman D, Wild AB, Zinser E, Sticht H, Muller YA, Steinkasserer A, Lechmann M. The CD83 Molecule - An Important Immune Checkpoint. Front Immunol 2020; 11:721. [PMID: 32362900 PMCID: PMC7181454 DOI: 10.3389/fimmu.2020.00721] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 03/30/2020] [Indexed: 12/19/2022] Open
Abstract
The CD83 molecule has been identified to be expressed on numerous activated immune cells, including B and T lymphocytes, monocytes, dendritic cells, microglia, and neutrophils. Both isoforms of CD83, the membrane-bound as well as its soluble form are topic of intensive research investigations. Several studies revealed that CD83 is not a typical co-stimulatory molecule, but rather plays a critical role in controlling and resolving immune responses. Moreover, CD83 is an essential factor during the differentiation of T and B lymphocytes, and the development and maintenance of tolerance. The identification of its interaction partners as well as signaling pathways have been an enigma for the last decades. Here, we report the latest data on the expression, structure, and the signaling partners of CD83. In addition, we review the regulatory functions of CD83, including its striking modulatory potential to maintain the balance between tolerance versus inflammation during homeostasis or pathologies. These immunomodulatory properties of CD83 emphasize its exceptional therapeutic potential, which has been documented in specific preclinical disease models.
Collapse
Affiliation(s)
- Linda Grosche
- Department of Immune Modulation, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Ilka Knippertz
- Department of Immune Modulation, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Christina König
- Department of Immune Modulation, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Dmytro Royzman
- Department of Immune Modulation, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Andreas B. Wild
- Department of Immune Modulation, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Elisabeth Zinser
- Department of Immune Modulation, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Heinrich Sticht
- Division of Bioinformatics, Institute of Biochemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Yves A. Muller
- Division of Biotechnology, Department of Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Alexander Steinkasserer
- Department of Immune Modulation, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Matthias Lechmann
- Department of Immune Modulation, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
8
|
Wild AB, Krzyzak L, Peckert K, Stich L, Kuhnt C, Butterhof A, Seitz C, Mattner J, Grüner N, Gänsbauer M, Purtak M, Soulat D, Winkler TH, Nitschke L, Zinser E, Steinkasserer A. CD83 orchestrates immunity toward self and non-self in dendritic cells. JCI Insight 2019; 4:e126246. [PMID: 31527313 PMCID: PMC6824307 DOI: 10.1172/jci.insight.126246] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 09/04/2019] [Indexed: 01/22/2023] Open
Abstract
Dendritic cells (DCs) are crucial to balance protective immunity and autoimmune inflammatory processes. Expression of CD83 is a well-established marker for mature DCs, although its physiological role is still not completely understood. Using a DC-specific CD83-conditional KO (CD83ΔDC) mouse, we provide new insights into the function of CD83 within this cell type. Interestingly, CD83-deficient DCs produced drastically increased IL-2 levels and displayed higher expression of the costimulatory molecules CD25 and OX40L, which causes superior induction of antigen-specific T cell responses and compromises Treg suppressive functions. This also directly translates into accelerated immune responses in vivo. Upon Salmonella typhimurium and Listeria monocytogenes infection, CD83ΔDC mice cleared both pathogens more efficiently, and CD83-deficient DCs expressed increased IL-12 levels after bacterial encounter. Using the experimental autoimmune encephalomyelitis model, autoimmune inflammation was dramatically aggravated in CD83ΔDC mice while resolution of inflammation was strongly reduced. This phenotype was associated with increased cell influx into the CNS accompanied by elevated Th17 cell numbers. Concomitantly, CD83ΔDC mice had reduced Treg numbers in peripheral lymphoid organs. In summary, we show that CD83 ablation on DCs results in enhanced immune responses by dysregulating tolerance mechanisms and thereby impairing resolution of inflammation, which also demonstrates high clinical relevance.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jochen Mattner
- Institute of Microbiology — Clinical Microbiology, Immunology and Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Niklas Grüner
- Institute of Microbiology — Clinical Microbiology, Immunology and Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Maximilian Gänsbauer
- Institute of Microbiology — Clinical Microbiology, Immunology and Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Martin Purtak
- Institute of Microbiology — Clinical Microbiology, Immunology and Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Didier Soulat
- Institute of Microbiology — Clinical Microbiology, Immunology and Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Thomas H. Winkler
- Division of Genetics, Department of Biology, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Lars Nitschke
- Division of Genetics, Department of Biology, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | | | | |
Collapse
|
9
|
Huo S, Zhang J, Liang S, Wu F, Zuo Y, Cui D, Zhang Y, Zhong Z, Zhong F. Membrane-bound and soluble porcine CD83 functions antithetically in T cell activation and dendritic cell differentiation in vitro. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2019; 99:103398. [PMID: 31121186 DOI: 10.1016/j.dci.2019.103398] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 05/16/2019] [Accepted: 05/16/2019] [Indexed: 06/09/2023]
Abstract
Emerging evidence suggests that CD83, a dendritic cells (DCs) maturation marker in humans and mice, may prossess immunomodulatory capacities. Although porcine CD83 shares ∼75% sequence homology with its human counterpart, whether it functions as an immunoregulatory molecule remains unknown. To investigate porcine CD83 function, we deleted it in porcine DCs by RNA intereference. Results show that membrane-bound CD83 (mCD83) promotes DC-mediated T cell proliferation and cytokine production, thus confirming its immunoregulatory capacity. Intriguingly, porcine soluble CD83 (sCD83) treatment instead led to inhibition of DC-mediated T cell activation. Moreover, porcine sCD83 also inhibited differentiation of prepheral blood mononuclear cells (PBMCs) into DCs. These results collectively indicate that in addition to being a DC maturation maker, both membrane bound and souble porcine CD83 serve as immunoregulatory molecules with opposite effects on DC-mediated T cell activation and DC differentiation.
Collapse
Affiliation(s)
- Shanshan Huo
- Laboratory of Molecular Virology and Immunology, College of Animal Science and Technology/College of Veterinary Medicine, Hebei Agricultural University, Baoding, Hebei, 071000, China; Hebei Engineering and Technology Research Center of Veterinary Biotechnology, Baoding, Hebei, 071000, China
| | - Jianlou Zhang
- Laboratory of Molecular Virology and Immunology, College of Animal Science and Technology/College of Veterinary Medicine, Hebei Agricultural University, Baoding, Hebei, 071000, China; Hebei Engineering and Technology Research Center of Veterinary Biotechnology, Baoding, Hebei, 071000, China
| | - Shuang Liang
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Fengyang Wu
- Laboratory of Molecular Virology and Immunology, College of Animal Science and Technology/College of Veterinary Medicine, Hebei Agricultural University, Baoding, Hebei, 071000, China; Hebei Engineering and Technology Research Center of Veterinary Biotechnology, Baoding, Hebei, 071000, China
| | - Yuzhu Zuo
- Laboratory of Molecular Virology and Immunology, College of Animal Science and Technology/College of Veterinary Medicine, Hebei Agricultural University, Baoding, Hebei, 071000, China; Hebei Engineering and Technology Research Center of Veterinary Biotechnology, Baoding, Hebei, 071000, China
| | - Dan Cui
- Laboratory of Molecular Virology and Immunology, College of Animal Science and Technology/College of Veterinary Medicine, Hebei Agricultural University, Baoding, Hebei, 071000, China; Hebei Engineering and Technology Research Center of Veterinary Biotechnology, Baoding, Hebei, 071000, China
| | - Yonghong Zhang
- Laboratory of Molecular Virology and Immunology, College of Animal Science and Technology/College of Veterinary Medicine, Hebei Agricultural University, Baoding, Hebei, 071000, China; Hebei Engineering and Technology Research Center of Veterinary Biotechnology, Baoding, Hebei, 071000, China
| | - Zhenyu Zhong
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| | - Fei Zhong
- Laboratory of Molecular Virology and Immunology, College of Animal Science and Technology/College of Veterinary Medicine, Hebei Agricultural University, Baoding, Hebei, 071000, China; Hebei Engineering and Technology Research Center of Veterinary Biotechnology, Baoding, Hebei, 071000, China.
| |
Collapse
|
10
|
Hos D, Matthaei M, Bock F, Maruyama K, Notara M, Clahsen T, Hou Y, Le VNH, Salabarria AC, Horstmann J, Bachmann BO, Cursiefen C. Immune reactions after modern lamellar (DALK, DSAEK, DMEK) versus conventional penetrating corneal transplantation. Prog Retin Eye Res 2019; 73:100768. [PMID: 31279005 DOI: 10.1016/j.preteyeres.2019.07.001] [Citation(s) in RCA: 173] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 07/01/2019] [Accepted: 07/02/2019] [Indexed: 12/12/2022]
Abstract
In the past decade, novel lamellar keratoplasty techniques such as Deep Anterior Lamellar Keratoplasty (DALK) for anterior keratoplasty and Descemet stripping automated endothelial keratoplasty (DSAEK)/Descemet membrane endothelial keratoplasty (DMEK) for posterior keratoplasty have been developed. DALK eliminates the possibility of endothelial allograft rejection, which is the main reason for graft failure after penetrating keratoplasty (PK). Compared to PK, the risk of endothelial graft rejection is significantly reduced after DSAEK/DMEK. Thus, with modern lamellar techniques, the clinical problem of endothelial graft rejection seems to be nearly solved in the low-risk situation. However, even with lamellar grafts there are epithelial, subepithelial and stromal immune reactions in DALK and endothelial immune reactions in DSAEK/DMEK, and not all keratoplasties can be performed in a lamellar fashion. Therefore, endothelial graft rejection in PK is still highly relevant, especially in the "high-risk" setting, where the cornea's (lymph)angiogenic and immune privilege is lost due to severe inflammation and pathological neovascularization. For these eyes, currently available treatment options are still unsatisfactory. In this review, we will describe currently used keratoplasty techniques, namely PK, DALK, DSAEK, and DMEK. We will summarize their indications, provide surgical descriptions, and comment on their complications and outcomes. Furthermore, we will give an overview on corneal transplant immunology. A specific focus will be placed on endothelial graft rejection and we will report on its incidence, clinical presentation, and current/future treatment and prevention options. Finally, we will speculate how the field of keratoplasty and prevention of corneal allograft rejection will develop in the future.
Collapse
Affiliation(s)
- Deniz Hos
- Department of Ophthalmology, University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Mario Matthaei
- Department of Ophthalmology, University of Cologne, Cologne, Germany
| | - Felix Bock
- Department of Ophthalmology, University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Kazuichi Maruyama
- Department of Innovative Visual Science, Graduate School of Medicine, Osaka University, Japan
| | - Maria Notara
- Department of Ophthalmology, University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Thomas Clahsen
- Department of Ophthalmology, University of Cologne, Cologne, Germany
| | - Yanhong Hou
- Department of Ophthalmology, University of Cologne, Cologne, Germany
| | - Viet Nhat Hung Le
- Department of Ophthalmology, University of Cologne, Cologne, Germany; Department of Ophthalmology, Hue College of Medicine and Pharmacy, Hue University, Viet Nam
| | | | - Jens Horstmann
- Department of Ophthalmology, University of Cologne, Cologne, Germany
| | - Bjoern O Bachmann
- Department of Ophthalmology, University of Cologne, Cologne, Germany
| | - Claus Cursiefen
- Department of Ophthalmology, University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.
| |
Collapse
|
11
|
Zinser E, Naumann R, Wild AB, Michalski J, Deinzer A, Stich L, Kuhnt C, Steinkasserer A, Knippertz I. Endogenous Expression of the Human CD83 Attenuates EAE Symptoms in Humanized Transgenic Mice and Increases the Activity of Regulatory T Cells. Front Immunol 2019; 10:1442. [PMID: 31293592 PMCID: PMC6603205 DOI: 10.3389/fimmu.2019.01442] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 06/07/2019] [Indexed: 12/17/2022] Open
Abstract
The CD83 is a type I membrane protein and part of the immunoglobulin superfamily of receptors. CD83 is involved in the regulation of antigen presentation and dendritic cell dependent allogeneic T cell proliferation. A soluble form of CD83 inhibits dendritic cell maturation and function. Furthermore, CD83 is expressed on activated B cells, T cells, and in particular on regulatory T cells. Previous studies on murine CD83 demonstrated this molecule to be involved in several immune-regulatory processes, comprising that CD83 plays a key role in the development und function of different immune cells. In order to get further insights into the function of the human CD83 and to provide preclinical tools to guide the function of CD83/sCD83 for therapeutic purposes we generated Bacterial Artificial Chromosomes (BAC) transgenic mice. BACs are excellent tools for manipulating large DNA fragments and are utilized to engineer transgenic mice by pronuclear injection. Two different founders of BAC transgenic mice expressing human CD83 (BAC-hCD83tg mice) were generated and were examined for the hCD83 expression on different immune cells as well as both the in vitro and in vivo role of human CD83 (hCD83) in health and disease. Here, we found the hCD83 molecule to be present on activated DCs, B cells and subtypes of CD4+ T cells. CD8+ T cells, on the other hand, showed almost no hCD83 expression. To address the function of hCD83, we performed in vitro mixed lymphocyte reactions (MLR) as well as suppression assays and we used the in vivo model of experimental autoimmune encephalomyelitis (EAE) comparing wild-type and hCD83-BAC mice. Results herein showed a clearly diminished capacity of hCD83-BAC-derived T cells to proliferate accompanied by an enhanced activation and suppressive activity of hCD83-BAC-derived Tregs. Furthermore, hCD83-BAC mice were found to recover faster from EAE-associated symptoms than wild-type mice, encouraging the relevance also of the hCD83 as a key molecule for the regulatory phenotype of Tregs in vitro and in vivo.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/immunology
- B-Lymphocytes/immunology
- B-Lymphocytes/pathology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/pathology
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Humans
- Immunoglobulins/genetics
- Immunoglobulins/immunology
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/immunology
- Mice
- Mice, Transgenic
- Somatostatin-Secreting Cells/immunology
- Somatostatin-Secreting Cells/pathology
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/pathology
- CD83 Antigen
Collapse
Affiliation(s)
- Elisabeth Zinser
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Ronald Naumann
- Transgenic Core Facility, Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Andreas B. Wild
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Julia Michalski
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Andrea Deinzer
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Lena Stich
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Christine Kuhnt
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| | | | - Ilka Knippertz
- Department of Immune Modulation, Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
12
|
Li Z, Ju X, Silveira PA, Abadir E, Hsu WH, Hart DNJ, Clark GJ. CD83: Activation Marker for Antigen Presenting Cells and Its Therapeutic Potential. Front Immunol 2019; 10:1312. [PMID: 31231400 PMCID: PMC6568190 DOI: 10.3389/fimmu.2019.01312] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 05/23/2019] [Indexed: 12/17/2022] Open
Abstract
CD83 is a member of the immunoglobulin (Ig) superfamily and is expressed in membrane bound or soluble forms. Membrane CD83 (mCD83) can be detected on a variety of activated immune cells, although it is most highly and stably expressed by mature dendritic cells (DC). mCD83 regulates maturation, activation and homeostasis. Soluble CD83 (sCD83), which is elevated in the serum of patients with autoimmune disease and some hematological malignancies is reported to have an immune suppressive function. While CD83 is emerging as a promising immune modulator with therapeutic potential, some important aspects such as its ligand/s, intracellular signaling pathways and modulators of its expression are unclear. In this review we discuss the recent biological findings and the potential clinical value of CD83 based therapeutics in various conditions including autoimmune disease, graft-vs.-host disease, transplantation and hematological malignancies.
Collapse
Affiliation(s)
- Ziduo Li
- Dendritic Cell Research, ANZAC Research Institute, Sydney, NSW, Australia
- Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Xinsheng Ju
- Dendritic Cell Research, ANZAC Research Institute, Sydney, NSW, Australia
- Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Pablo A. Silveira
- Dendritic Cell Research, ANZAC Research Institute, Sydney, NSW, Australia
- Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Edward Abadir
- Dendritic Cell Research, ANZAC Research Institute, Sydney, NSW, Australia
- Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Wei-Hsun Hsu
- Dendritic Cell Research, ANZAC Research Institute, Sydney, NSW, Australia
- Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Derek N. J. Hart
- Dendritic Cell Research, ANZAC Research Institute, Sydney, NSW, Australia
- Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Georgina J. Clark
- Dendritic Cell Research, ANZAC Research Institute, Sydney, NSW, Australia
- Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
13
|
Huo S, Zhang J, Wu F, Zuo Y, Cui D, Li X, Zhong Z, Zhong F. Porcine CD83 is a glycosylated dimeric protein existing naturally in membrane-bound and soluble forms. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2019; 90:60-69. [PMID: 30193829 DOI: 10.1016/j.dci.2018.09.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 09/03/2018] [Accepted: 09/03/2018] [Indexed: 06/08/2023]
Abstract
Human and mouse CD83 have been well characteized, however, the other mammalian CD83 genes have not been cloned and characterized. In this study, the porcine CD83 (pCD83) was cloned, expressed and characterized, and showed that the pCD83 gene has 81% and 74% homologies with humans and mice, respectively, which was identified to be glycosylated when expressed in eukaryotic cells, existing naturally in two forms: membrance-bound CD83 (mCD83) and soluble CD83 (sCD83), the latter was identified to be generated mainly from mCD83 by proteolytic shedding. The pCD83 was a dimmer mediated by intermolecular disulfide bond formed by the fifth cysteine in the exrtracellular domain. Functionally, the recombinant porcine sCD83 was preliminarily tested to have the ability to inhibit DC-mediated T cell activition. This study provided necessary fundation for further investigation on pCD83 functions.
Collapse
Affiliation(s)
- Shanshan Huo
- Laboratory of Molecular Virology and Immunology, College of Animal Science and Technology/College of Veterinary Medicine, Agricultural University of Hebei, Baoding, Hebei, 071000, China; Hebei Engineering and Technology Research Center of Veterinary Biotechnology, Baoding, Hebei, 071000, China
| | - Jianlou Zhang
- Laboratory of Molecular Virology and Immunology, College of Animal Science and Technology/College of Veterinary Medicine, Agricultural University of Hebei, Baoding, Hebei, 071000, China; Hebei Engineering and Technology Research Center of Veterinary Biotechnology, Baoding, Hebei, 071000, China
| | - Fengyang Wu
- Laboratory of Molecular Virology and Immunology, College of Animal Science and Technology/College of Veterinary Medicine, Agricultural University of Hebei, Baoding, Hebei, 071000, China
| | - Yuzhu Zuo
- Laboratory of Molecular Virology and Immunology, College of Animal Science and Technology/College of Veterinary Medicine, Agricultural University of Hebei, Baoding, Hebei, 071000, China; Hebei Engineering and Technology Research Center of Veterinary Biotechnology, Baoding, Hebei, 071000, China
| | - Dan Cui
- Laboratory of Molecular Virology and Immunology, College of Animal Science and Technology/College of Veterinary Medicine, Agricultural University of Hebei, Baoding, Hebei, 071000, China; Hebei Engineering and Technology Research Center of Veterinary Biotechnology, Baoding, Hebei, 071000, China
| | - Xiujin Li
- Department of Biotechnology, College of Environmental and Chemical Engineering, Yanshan University, Qinhuangdao, Hebei, 066004, China
| | - Zhenyu Zhong
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| | - Fei Zhong
- Laboratory of Molecular Virology and Immunology, College of Animal Science and Technology/College of Veterinary Medicine, Agricultural University of Hebei, Baoding, Hebei, 071000, China; Hebei Engineering and Technology Research Center of Veterinary Biotechnology, Baoding, Hebei, 071000, China.
| |
Collapse
|
14
|
Bo L, Guojun T, Li G. An Expanded Neuroimmunomodulation Axis: sCD83-Indoleamine 2,3-Dioxygenase-Kynurenine Pathway and Updates of Kynurenine Pathway in Neurologic Diseases. Front Immunol 2018; 9:1363. [PMID: 29963055 PMCID: PMC6013554 DOI: 10.3389/fimmu.2018.01363] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 06/01/2018] [Indexed: 12/30/2022] Open
Abstract
Many neurologic diseases are related to autoimmune dysfunction and a variety of molecules or reaction pathways are involved in the regulation of immune function of the nervous system. Soluble CD83 (sCD83) is the soluble form of CD83, a specific marker of mature dendritic cell, which has recently been shown to have an immunomodulatory effect. Indoleamine 2,3-dioxygenase (IDO; corresponding enzyme intrahepatic, tryptophan 2,3-dioxygenase, TDO), a rate-limiting enzyme of extrahepatic tryptophan kynurenine pathway (KP) participates in the immunoregulation through a variety of mechanisms solely or with the synergy of sCD83, and the imbalances of metabolites of KP were associated with immune dysfunction. With the complement of sCD83 to IDO-KP, a previously known immunomodulatory axis, this review focused on an expanded neuroimmunomodulation axis: sCD83-IDO-KP and its involvement in nervous system diseases.
Collapse
Affiliation(s)
- Li Bo
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Tan Guojun
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Guo Li
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
15
|
Doebbeler M, Koenig C, Krzyzak L, Seitz C, Wild A, Ulas T, Baßler K, Kopelyanskiy D, Butterhof A, Kuhnt C, Kreiser S, Stich L, Zinser E, Knippertz I, Wirtz S, Riegel C, Hoffmann P, Edinger M, Nitschke L, Winkler T, Schultze JL, Steinkasserer A, Lechmann M. CD83 expression is essential for Treg cell differentiation and stability. JCI Insight 2018; 3:99712. [PMID: 29875316 PMCID: PMC6124443 DOI: 10.1172/jci.insight.99712] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 05/01/2018] [Indexed: 12/21/2022] Open
Abstract
Foxp3-positive regulatory T cells (Tregs) are crucial for the maintenance of immune homeostasis and keep immune responses in check. Upon activation, Tregs are transferred into an effector state expressing transcripts essential for their suppressive activity, migration, and survival. However, it is not completely understood how different intrinsic and environmental factors control differentiation. Here, we present for the first time to our knowledge data suggesting that Treg-intrinsic expression of CD83 is essential for Treg differentiation upon activation. Interestingly, mice with Treg-intrinsic CD83 deficiency are characterized by a proinflammatory phenotype. Furthermore, the loss of CD83 expression by Tregs leads to the downregulation of Treg-specific differentiation markers and the induction of an inflammatory profile. In addition, Treg-specific conditional knockout mice showed aggravated autoimmunity and an impaired resolution of inflammation. Altogether, our results show that CD83 expression in Tregs is an essential factor for the development and function of effector Tregs upon activation. Since Tregs play a crucial role in the maintenance of immune tolerance and thus prevention of autoimmune disorders, our findings are also clinically relevant.
Collapse
Affiliation(s)
- Marina Doebbeler
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | - Christina Koenig
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | - Lena Krzyzak
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | - Christine Seitz
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | - Andreas Wild
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | - Thomas Ulas
- Genomics & Immunoregulation, LIMES-Institute, University of Bonn, Bonn, Germany
| | - Kevin Baßler
- Genomics & Immunoregulation, LIMES-Institute, University of Bonn, Bonn, Germany
| | - Dmitry Kopelyanskiy
- Genomics & Immunoregulation, LIMES-Institute, University of Bonn, Bonn, Germany
| | - Alina Butterhof
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | - Christine Kuhnt
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | - Simon Kreiser
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | - Lena Stich
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | - Elisabeth Zinser
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | - Ilka Knippertz
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | - Stefan Wirtz
- Department of Medicine 1, University Hospital Erlangen, Erlangen, Germany
| | - Christin Riegel
- Department of Internal Medicine 3, University Hospital Regensburg, Regensburg, Germany
| | - Petra Hoffmann
- Department of Internal Medicine 3, University Hospital Regensburg, Regensburg, Germany
| | - Matthias Edinger
- Department of Internal Medicine 3, University Hospital Regensburg, Regensburg, Germany
| | - Lars Nitschke
- Division of Genetics, Department of Biology, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Thomas Winkler
- Division of Genetics, Department of Biology, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Joachim L. Schultze
- Genomics & Immunoregulation, LIMES-Institute, University of Bonn, Bonn, Germany
| | - Alexander Steinkasserer
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | - Matthias Lechmann
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| |
Collapse
|
16
|
Vendelova E, Ashour D, Blank P, Erhard F, Saliba AE, Kalinke U, Lutz MB. Tolerogenic Transcriptional Signatures of Steady-State and Pathogen-Induced Dendritic Cells. Front Immunol 2018. [PMID: 29541071 PMCID: PMC5835767 DOI: 10.3389/fimmu.2018.00333] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Dendritic cells (DCs) are key directors of tolerogenic and immunogenic immune responses. During the steady state, DCs maintain T cell tolerance to self-antigens by multiple mechanisms including inducing anergy, deletion, and Treg activity. All of these mechanisms help to prevent autoimmune diseases or other hyperreactivities. Different DC subsets contribute to pathogen recognition by expression of different subsets of pattern recognition receptors, including Toll-like receptors or C-type lectins. In addition to the triggering of immune responses in infected hosts, most pathogens have evolved mechanisms for evasion of targeted responses. One such strategy is characterized by adopting the host’s T cell tolerance mechanisms. Understanding these tolerogenic mechanisms is of utmost importance for therapeutic approaches to treat immune pathologies, tumors and infections. Transcriptional profiling has developed into a potent tool for DC subset identification. Here, we review and compile pathogen-induced tolerogenic transcriptional signatures from mRNA profiling data of currently available bacterial- or helminth-induced transcriptional signatures. We compare them with signatures of tolerogenic steady-state DC subtypes to identify common and divergent strategies of pathogen induced immune evasion. Candidate molecules are discussed in detail. Our analysis provides further insights into tolerogenic DC signatures and their exploitation by different pathogens.
Collapse
Affiliation(s)
- Emilia Vendelova
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Diyaaeldin Ashour
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Patrick Blank
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, Germany
| | - Florian Erhard
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | | | - Ulrich Kalinke
- Institute for Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research and the Hannover Medical School, Hannover, Germany
| | - Manfred B Lutz
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
17
|
Heilingloh CS, Klingl S, Egerer-Sieber C, Schmid B, Weiler S, Mühl-Zürbes P, Hofmann J, Stump JD, Sticht H, Kummer M, Steinkasserer A, Muller YA. Crystal Structure of the Extracellular Domain of the Human Dendritic Cell Surface Marker CD83. J Mol Biol 2017; 429:1227-1243. [PMID: 28315353 DOI: 10.1016/j.jmb.2017.03.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Revised: 02/10/2017] [Accepted: 03/05/2017] [Indexed: 11/27/2022]
Abstract
CD83 is a type-I membrane protein and an efficient marker for identifying mature dendritic cells. Whereas membrane-bound, full-length CD83 co-stimulates the immune system, a soluble variant (sCD83), consisting of the extracellular domain only, displays strong immune-suppressive activities. Besides a prediction that sCD83 adopts a V-set Ig-like fold, however, little is known about the molecular architecture of CD83 and the mechanism by which CD83 exerts its function on dendritic cells and additional immune cells. Here, we report the crystal structure of human sCD83 up to a resolution of 1.7Å solved in three different crystal forms. Interestingly, β-strands C', C″, and D that are typical for V-set Ig-domains could not be traced in sCD83. Mass spectrometry analyses, limited proteolysis experiments, and bioinformatics studies show that the corresponding segment displays enhanced main-chain accessibility, extraordinary low sequence conservation, and a predicted high disorder propensity. Chimeric proteins with amino acid swaps in this segment show unaltered immune-suppressive activities in a TNF-α assay when compared to wild-type sCD83. This strongly indicates that this segment does not participate in the biological activity of CD83. The crystal structure of CD83 shows the recurrent formation of dimers and trimers in the various crystal forms and reveals strong structural similarities between sCD83 and B7 family members and CD48, a signaling lymphocyte activation molecule family member. This suggests that CD83 exerts its immunological activity by mixed homotypic and heterotypic interactions as typically observed for proteins present in the immunological synapse.
Collapse
Affiliation(s)
- Christiane S Heilingloh
- Department of Immune Modulation, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Hartmannstr. 14, D-91052 Erlangen, Germany
| | - Stefan Klingl
- Division of Biotechnology, Department of Biology, Friedrich-Alexander-University Erlangen-Nürnberg, Henkestr. 91, D-91052 Erlangen, Germany
| | - Claudia Egerer-Sieber
- Division of Biotechnology, Department of Biology, Friedrich-Alexander-University Erlangen-Nürnberg, Henkestr. 91, D-91052 Erlangen, Germany
| | - Benedikt Schmid
- Division of Biotechnology, Department of Biology, Friedrich-Alexander-University Erlangen-Nürnberg, Henkestr. 91, D-91052 Erlangen, Germany
| | - Sigrid Weiler
- Division of Biotechnology, Department of Biology, Friedrich-Alexander-University Erlangen-Nürnberg, Henkestr. 91, D-91052 Erlangen, Germany
| | - Petra Mühl-Zürbes
- Department of Immune Modulation, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Hartmannstr. 14, D-91052 Erlangen, Germany
| | - Jörg Hofmann
- Division of Biochemistry, Department of Biology, Friedrich-Alexander-University Erlangen-Nürnberg, Staudtstr. 5, D-91058 Erlangen, Germany
| | - Joachim D Stump
- Division of Bioinformatics, Institute of Biochemistry, Friedrich-Alexander-University Erlangen-Nürnberg, Fahrstraße 17, D-91054 Erlangen, Germany
| | - Heinrich Sticht
- Division of Bioinformatics, Institute of Biochemistry, Friedrich-Alexander-University Erlangen-Nürnberg, Fahrstraße 17, D-91054 Erlangen, Germany
| | - Mirko Kummer
- Department of Immune Modulation, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Hartmannstr. 14, D-91052 Erlangen, Germany
| | - Alexander Steinkasserer
- Department of Immune Modulation, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Hartmannstr. 14, D-91052 Erlangen, Germany
| | - Yves A Muller
- Division of Biotechnology, Department of Biology, Friedrich-Alexander-University Erlangen-Nürnberg, Henkestr. 91, D-91052 Erlangen, Germany.
| |
Collapse
|
18
|
Horvatinovich JM, Grogan EW, Norris M, Steinkasserer A, Lemos H, Mellor AL, Tcherepanova IY, Nicolette CA, DeBenedette MA. Soluble CD83 Inhibits T Cell Activation by Binding to the TLR4/MD-2 Complex on CD14 + Monocytes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2017; 198:2286-2301. [PMID: 28193829 PMCID: PMC5337811 DOI: 10.4049/jimmunol.1600802] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 01/13/2017] [Indexed: 12/17/2022]
Abstract
The transmembrane protein CD83, expressed on APCs, B cells, and T cells, can be expressed as a soluble form generated by alternative splice variants and/or by shedding. Soluble CD83 (sCD83) was shown to be involved in negatively regulating the immune response. sCD83 inhibits T cell proliferation in vitro, supports allograft survival in vivo, prevents corneal transplant rejection, and attenuates the progression and severity of autoimmune diseases and experimental colitis. Although sCD83 binds to human PBMCs, the specific molecules that bind sCD83 have not been identified. In this article, we identify myeloid differentiation factor-2 (MD-2), the coreceptor within the TLR4/MD-2 receptor complex, as the high-affinity sCD83 binding partner. TLR4/MD-2 mediates proinflammatory signal delivery following recognition of bacterial LPSs. However, altering TLR4 signaling can attenuate the proinflammatory cascade, leading to LPS tolerance. Our data show that binding of sCD83 to MD-2 alters this signaling cascade by rapidly degrading IL-1R-associated kinase-1, leading to induction of the anti-inflammatory mediators IDO, IL-10, and PGE2 in a COX-2-dependent manner. sCD83 inhibited T cell proliferation, blocked IL-2 secretion, and rendered T cells unresponsive to further downstream differentiation signals mediated by IL-2. Therefore, we propose the tolerogenic mechanism of action of sCD83 to be dependent on initial interaction with APCs, altering early cytokine signal pathways and leading to T cell unresponsiveness.
Collapse
Affiliation(s)
| | | | - Marcus Norris
- Research Department, Argos Therapeutics, Inc., Durham, NC 27704
| | - Alexander Steinkasserer
- Cancer Immunology, Department of Immune Modulation, University Hospital Erlangen, University of Erlangen-Nuremberg, D-91052 Erlangen, Germany; and
| | - Henrique Lemos
- Inflammation and Tolerance Program, Cancer Center, Georgia Regents University, Augusta, GA 30912
| | - Andrew L Mellor
- Inflammation and Tolerance Program, Cancer Center, Georgia Regents University, Augusta, GA 30912
| | | | | | | |
Collapse
|
19
|
Heilingloh CS, Grosche L, Kummer M, Mühl-Zürbes P, Kamm L, Scherer M, Latzko M, Stamminger T, Steinkasserer A. The Major Immediate-Early Protein IE2 of Human Cytomegalovirus Is Sufficient to Induce Proteasomal Degradation of CD83 on Mature Dendritic Cells. Front Microbiol 2017; 8:119. [PMID: 28203230 PMCID: PMC5285329 DOI: 10.3389/fmicb.2017.00119] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 01/17/2017] [Indexed: 01/03/2023] Open
Abstract
Human cytomegalovirus (HCMV) is the prototypic beta-herpesvirus and widespread throughout the human population. While infection is asymptomatic in healthy individuals, it can lead to high morbidity and mortality in immunocompromised persons. Importantly, HCMV evolved multiple strategies to interfere with immune cell function in order to establish latency in infected individuals. As mature DCs (mDCs) are antigen-presenting cells able to activate naïve T cells they play a crucial role during induction of effective antiviral immune responses. Interestingly, earlier studies demonstrated that the functionally important mDC surface molecule CD83 is down-regulated upon HCMV infection resulting in a reduced T cell stimulatory capacity of the infected cells. However, the viral effector protein and the precise mechanism of HCMV-mediated CD83 reduction remain to be discovered. Using flow cytometric analyses, we observed significant down-modulation of CD83 surface expression becoming significant already 12 h after HCMV infection. Moreover, Western bot analyses revealed that, in sharp contrast to previous studies, loss of CD83 is not restricted to the membrane-bound molecule, but also occurs intracellularly. Furthermore, inhibition of the proteasome almost completely restored CD83 surface expression during HCMV infection. Results of infection kinetics and cycloheximide-actinomycin D-chase experiments, strongly suggested that an HCMV immediate early gene product is responsible for the induction of CD83 down-modulation. Consequently, we were able to identify the major immediate early protein IE2 as the viral effector protein that induces proteasomal CD83 degradation.
Collapse
Affiliation(s)
| | - Linda Grosche
- Department of Immune Modulation, University Hospital Erlangen Erlangen, Germany
| | - Mirko Kummer
- Department of Immune Modulation, University Hospital Erlangen Erlangen, Germany
| | - Petra Mühl-Zürbes
- Department of Immune Modulation, University Hospital Erlangen Erlangen, Germany
| | - Lisa Kamm
- Department of Immune Modulation, University Hospital Erlangen Erlangen, Germany
| | - Myriam Scherer
- Institute for Clinical and Molecular Virology, University of Erlangen-Nuremberg Erlangen, Germany
| | - Melanie Latzko
- Department of Immune Modulation, University Hospital Erlangen Erlangen, Germany
| | - Thomas Stamminger
- Institute for Clinical and Molecular Virology, University of Erlangen-Nuremberg Erlangen, Germany
| | | |
Collapse
|
20
|
Ju X, Silveira PA, Hsu WH, Elgundi Z, Alingcastre R, Verma ND, Fromm PD, Hsu JL, Bryant C, Li Z, Kupresanin F, Lo TH, Clarke C, Lee K, McGuire H, Fazekas de St Groth B, Larsen SR, Gibson J, Bradstock KF, Clark GJ, Hart DNJ. The Analysis of CD83 Expression on Human Immune Cells Identifies a Unique CD83+-Activated T Cell Population. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 197:4613-4625. [PMID: 27837105 DOI: 10.4049/jimmunol.1600339] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 10/10/2016] [Indexed: 02/02/2023]
Abstract
CD83 is a member of the Ig gene superfamily, first identified in activated lymphocytes. Since then, CD83 has become an important marker for defining activated human dendritic cells (DC). Several potential CD83 mRNA isoforms have been described, including a soluble form detected in human serum, which may have an immunosuppressive function. To further understand the biology of CD83, we examined its expression in different human immune cell types before and after activation using a panel of mouse and human anti-human CD83 mAb. The mouse anti-human CD83 mAbs, HB15a and HB15e, and the human anti-human CD83 mAb, 3C12C, were selected to examine cytoplasmic and surface CD83 expression, based on their different binding characteristics. Glycosylation of CD83, the CD83 mRNA isoforms, and soluble CD83 released differed among blood DC, monocytes, and monocyte-derived DC, and other immune cell types. A small T cell population expressing surface CD83 was identified upon T cell stimulation and during allogeneic MLR. This subpopulation appeared specifically during viral Ag challenge. We did not observe human CD83 on unstimulated human natural regulatory T cells (Treg), in contrast to reports describing expression of CD83 on mouse Treg. CD83 expression was increased on CD4+, CD8+ T, and Treg cells in association with clinical acute graft-versus-host disease in allogeneic hematopoietic cell transplant recipients. The differential expression and function of CD83 on human immune cells reveal potential new roles for this molecule as a target of therapeutic manipulation in transplantation, inflammation, and autoimmune diseases.
Collapse
Affiliation(s)
- Xinsheng Ju
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, New South Wales 2139, Australia
| | - Pablo A Silveira
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, New South Wales 2139, Australia
- Sydney Medical School, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Wei-Hsun Hsu
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, New South Wales 2139, Australia
- Sydney Medical School, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Zehra Elgundi
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, New South Wales 2139, Australia
| | - Renz Alingcastre
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, New South Wales 2139, Australia
| | - Nirupama D Verma
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, New South Wales 2139, Australia
| | - Phillip D Fromm
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, New South Wales 2139, Australia
- Sydney Medical School, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Jennifer L Hsu
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, New South Wales 2139, Australia
- Institute of Haematology, Royal Prince Alfred Hospital, University of Sydney, Sydney, New South Wales 2050, Australia
| | - Christian Bryant
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, New South Wales 2139, Australia
- Institute of Haematology, Royal Prince Alfred Hospital, University of Sydney, Sydney, New South Wales 2050, Australia
| | - Ziduo Li
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, New South Wales 2139, Australia
- Sydney Medical School, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Fiona Kupresanin
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, New South Wales 2139, Australia
| | - Tsun-Ho Lo
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, New South Wales 2139, Australia
- Sydney Medical School, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Candice Clarke
- Anatomical Pathology Department, Concord Repatriation General Hospital, Sydney, New South Wales 2139, Australia; and
| | - Kenneth Lee
- Sydney Medical School, University of Sydney, Sydney, New South Wales 2006, Australia
- Anatomical Pathology Department, Concord Repatriation General Hospital, Sydney, New South Wales 2139, Australia; and
| | - Helen McGuire
- Centenary Institute, Royal Prince Alfred Hospital, Sydney, New South Wales 2050, Australia
| | | | - Stephen R Larsen
- Sydney Medical School, University of Sydney, Sydney, New South Wales 2006, Australia
- Institute of Haematology, Royal Prince Alfred Hospital, University of Sydney, Sydney, New South Wales 2050, Australia
| | - John Gibson
- Sydney Medical School, University of Sydney, Sydney, New South Wales 2006, Australia
- Institute of Haematology, Royal Prince Alfred Hospital, University of Sydney, Sydney, New South Wales 2050, Australia
| | - Kenneth F Bradstock
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, New South Wales 2139, Australia
- Sydney Medical School, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Georgina J Clark
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, New South Wales 2139, Australia
- Sydney Medical School, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Derek N J Hart
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, New South Wales 2139, Australia;
- Sydney Medical School, University of Sydney, Sydney, New South Wales 2006, Australia
| |
Collapse
|
21
|
Heilingloh CS, Kummer M, Mühl-Zürbes P, Drassner C, Daniel C, Klewer M, Steinkasserer A. L Particles Transmit Viral Proteins from Herpes Simplex Virus 1-Infected Mature Dendritic Cells to Uninfected Bystander Cells, Inducing CD83 Downmodulation. J Virol 2015; 89:11046-55. [PMID: 26311871 PMCID: PMC4621140 DOI: 10.1128/jvi.01517-15] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 08/19/2015] [Indexed: 01/11/2023] Open
Abstract
UNLABELLED Mature dendritic cells (mDCs) are known as the most potent antigen-presenting cells (APCs) since they are also able to prime/induce naive T cells. Thus, mDCs play a pivotal role during the induction of antiviral immune responses. Remarkably, the cell surface molecule CD83, which was shown to have costimulatory properties, is targeted by herpes simplex virus 1 (HSV-1) for viral immune escape. Infection of mDCs with HSV-1 results in downmodulation of CD83, resulting in reduced T cell stimulation. In this study, we report that not only infected mDCs but also uninfected bystander cells in an infected culture show a significant CD83 reduction. We demonstrate that this effect is independent of phagocytosis and transmissible from infected to uninfected mDCs. The presence of specific viral proteins found in these uninfected bystander cells led to the hypothesis that viral proteins are transferred from infected to uninfected cells via L particles. These L particles are generated during lytic replication in parallel with full virions, called H particles. L particles contain viral proteins but lack the viral capsid and DNA. Therefore, these particles are not infectious but are able to transfer several viral proteins. Incubation of mDCs with L particles indeed reduced CD83 expression on uninfected bystander DCs, providing for the first time evidence that functional viral proteins are transmitted via L particles from infected mDCs to uninfected bystander cells, thereby inducing CD83 downmodulation. IMPORTANCE HSV-1 has evolved a number of strategies to evade the host's immune system. Among others, HSV-1 infection of mDCs results in an inhibited T cell activation caused by degradation of CD83. Interestingly, CD83 is lost not only from HSV-1-infected mDCs but also from uninfected bystander cells. The release of so-called L particles, which contain several viral proteins but lack capsid and DNA, during infection is a common phenomenon observed among several viruses, such as human cytomegalovirus (HCMV), Epstein-Barr virus, and HSV-1. However, the detailed function of these particles is poorly understood. Here, we provide for the first time evidence that functional viral proteins can be transferred to uninfected bystander mDCs via L particles, revealing important biological functions of these particles during lytic replication. Therefore, the transfer of viral proteins by L particles to modulate uninfected bystander cells may represent an additional strategy for viral immune escape.
Collapse
Affiliation(s)
| | - Mirko Kummer
- Department of Immune Modulation, University Hospital Erlangen, Erlangen, Germany
| | - Petra Mühl-Zürbes
- Department of Immune Modulation, University Hospital Erlangen, Erlangen, Germany
| | - Christina Drassner
- Department of Immune Modulation, University Hospital Erlangen, Erlangen, Germany
| | - Christoph Daniel
- Department of Pathology, Nephropathology, University Hospital Erlangen, Erlangen, Germany
| | - Monika Klewer
- Department of Pathology, Nephropathology, University Hospital Erlangen, Erlangen, Germany
| | | |
Collapse
|
22
|
Kreiser S, Eckhardt J, Kuhnt C, Stein M, Krzyzak L, Seitz C, Tucher C, Knippertz I, Becker C, Günther C, Steinkasserer A, Lechmann M. Murine CD83-positive T cells mediate suppressor functions in vitro and in vivo. Immunobiology 2015; 220:270-9. [PMID: 25151500 DOI: 10.1016/j.imbio.2014.08.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 07/22/2014] [Accepted: 08/04/2014] [Indexed: 10/24/2022]
Abstract
The CD83 molecule (CD83) is a well-known surface marker present on mature dendritic cells (mDC). In this study, we show that CD83 is also expressed on a subset of T cells which mediate regulatory T cell (Treg)-like suppressor functions in vitro and in vivo. Treg-associated molecules including CD25, cytotoxic T lymphocyte antigen-4 (CTLA-4), glucocorticoid-induced TNFR family-related gene (GITR), Helios and neuropilin-1 (NRP-1) as well as forkhead box protein 3 (FOXP3) were specifically expressed by these CD83(+) T cells. In contrast, CD83(-) T cells showed a naive T cell phenotype with effector T cell properties upon activation. Noteworthy, CD83(-) T cells were not able to upregulate CD83 despite activation. Furthermore, CD83(+) T cells suppressed the proliferation and inflammatory cytokine release of CD83(-) T cells in vitro. Strikingly, stimulated CD83(+) T cells released soluble CD83 (sCD83), which has been reported to possess immunosuppressive properties. In vivo, using the murine transfer colitis model we could show that CD83(+) T cells were able to suppress colitis symptoms while CD83(-) T cells possessed effector functions. In addition, this CD83 expression is also conserved on expanded human Treg. Thus, from these studies we conclude that CD83(+) T cells share important features with regulatory T cells, identifying CD83 as a novel lineage marker to discriminate between different T cell populations.
Collapse
Affiliation(s)
- Simon Kreiser
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen D-91052, Germany
| | - Jenny Eckhardt
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen D-91052, Germany
| | - Christine Kuhnt
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen D-91052, Germany
| | - Marcello Stein
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen D-91052, Germany
| | - Lena Krzyzak
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen D-91052, Germany
| | - Christine Seitz
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen D-91052, Germany
| | - Christine Tucher
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen D-91052, Germany
| | - Ilka Knippertz
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen D-91052, Germany
| | - Christoph Becker
- Department of Medicine 1, University Hospital Erlangen, Erlangen D-91052, Germany
| | - Claudia Günther
- Department of Medicine 1, University Hospital Erlangen, Erlangen D-91052, Germany
| | - Alexander Steinkasserer
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen D-91052, Germany
| | - Matthias Lechmann
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen D-91052, Germany; Department of Medicine 1, University Hospital Erlangen, Erlangen D-91052, Germany.
| |
Collapse
|
23
|
Yang Y, Xin Z, Chu J, Li N, Sun T. Involvement of Caveolin-1 in CD83 Internalization in Mouse Dendritic Cells. Cell Transplant 2014; 24:1395-404. [PMID: 24898475 DOI: 10.3727/096368914x682116] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
To become potent T-cell stimulators, DCs need to mature. Treatment with soluble CD83 (sCD83) induces immune tolerance and protects against transplant rejection by maintaining dendritic cells in an immature, tolerogenic state. Until now, the mechanism through which sCD83 keeps DCs immature has not been investigated. The internalizing pathway of CD83 was screened by Western blot, and the direct interactions between internalized proteins were verified through coimmunoprecipitation (co-IP) and transmission electron microscopy (TEM). CD83 plasma membrane levels were detected by Western blot using a plasma membrane protein extraction protocol. The changes in CD83 surface levels in DCs were detected by flow cytometry. Caveolin-1 function was detected in a kidney transplant model. In this study, we demonstrated that caveolin-1 could affect CD83 level during endocytosis in mouse DCs. Caveolin-1 coprecipitates with CD83, as demonstrated by co-IP analysis. TEM morphometric analysis of the entire CD83 distribution associated with internalized caveolin-1 demonstrated a significant interaction in cellular vesicles. sCD83 reduces endogenous CD83 plasma membrane levels, and caveolin-1 knockdown reverts CD83 levels in plasma membrane. sCD83 treatment decreases CD83 surface levels in DCs. siRNA to caveolin-1 in DCs inhibits this effect of sCD83. The effects of sCD83-treated DCs were proved in CD1 mice. Knocking down caveolin-1 in DCs obstructs the effects of sCD83 on kidney transplant. In conclusion, our data indicated that a caveolin-dependent endocytic pathway is involved in CD83 internalization in DCs and that caveolin-1 is involved in the activity of DCs.
Collapse
Affiliation(s)
- Yuejing Yang
- The 2nd Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | | | | | | | | |
Collapse
|
24
|
Heilingloh CS, Mühl-Zürbes P, Steinkasserer A, Kummer M. Herpes simplex virus type 1 ICP0 induces CD83 degradation in mature dendritic cells independent of its E3 ubiquitin ligase function. J Gen Virol 2014; 95:1366-1375. [PMID: 24643878 DOI: 10.1099/vir.0.062810-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Mature dendritic cells (mDCs) are the most potent antigen-presenting cells known today, as they are the only antigen-presenting cells able to induce naïve T-cells. Therefore, they play a crucial role during the induction of effective antiviral immune responses. Interestingly, the surface molecule CD83 expressed on mDCs is targeted by several viruses. As CD83 has been shown to exert co-stimulatory functions on mDCs, its downmodulation represents a viral immune escape mechanism. Mechanistically, it has been shown that herpes simplex virus type 1 infection leads to proteasomal degradation of CD83, resulting in a strongly diminished T-cell stimulatory capacity of the infected mDC. Previous data suggest that the viral immediate-early protein ICP0 (infected-cell protein 0) plays an important role in this process. In the present study, we showed that ICP0 is sufficient to induce CD83 degradation in the absence of any other viral factor. However, the mechanism of ICP0-mediated CD83 degradation is not yet understood. Here, we provide evidence that ubiquitination of lysine residues is, despite the published E3 ubiquitin ligase activity of ICP0, not necessary for CD83 degradation. This finding was underlined by the observation that expression of an ICP0 mutant lacking the E3 ubiquitin ligase domain in mDCs still induced CD83 degradation. Finally, inhibition of E1 activating enzyme using the specific inhibitor 4[4-(5-nitro-furan-2-ylmethylene)-3.5-dioxo-pyrazolidin-1-yl]-benzoic acid ethyl ester did not prevent CD83 degradation. Taken together, our data provide strong evidence that ICP0 alone induces CD83 degradation independent of its E3 ubiquitin ligase function and of the ubiquitin machinery.
Collapse
Affiliation(s)
- Christiane S Heilingloh
- Department of Immune Modulation, University Hospital Erlangen, Hartmannstrasse 14, D-91052 Erlangen, Germany
| | - Petra Mühl-Zürbes
- Department of Immune Modulation, University Hospital Erlangen, Hartmannstrasse 14, D-91052 Erlangen, Germany
| | - Alexander Steinkasserer
- Department of Immune Modulation, University Hospital Erlangen, Hartmannstrasse 14, D-91052 Erlangen, Germany
| | - Mirko Kummer
- Department of Immune Modulation, University Hospital Erlangen, Hartmannstrasse 14, D-91052 Erlangen, Germany
| |
Collapse
|
25
|
Pinho MP, Migliori IK, Flatow EA, Barbuto JAM. Dendritic cell membrane CD83 enhances immune responses by boosting intracellular calcium release in T lymphocytes. J Leukoc Biol 2014; 95:755-762. [PMID: 24436459 DOI: 10.1189/jlb.0413239] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 12/03/2013] [Accepted: 12/27/2013] [Indexed: 12/22/2022] Open
Abstract
CD83 is a marker of mDCs directly related to their lymphostimulatory ability. Some data suggest that it has a central role in the immune system regulation, but how this function is performed remains to be determined. This work aimed to analyze the influence of CD83, present in mDCs, in the modulation of calcium signaling in T lymphocytes. Mo were differentiated into iDCs and activated with TNF-α. iDCs were treated, 4 h before activation, with siRNACD83, to reduce CD83 expression. Purified allogeneic T lymphocytes were labeled with the calcium indicator Fluo-4-AM, and calcium mobilization in the presence of mDCs was analyzed. CD83 knockdown mDCs induced lower calcium signal amplitude in T lymphocytes (29.0±10.0) compared with siRNAscr-treated mDCs (45.5±5.3). In another set of experiments, surface mDC CD83 was blocked with a specific mAb, and again, decreased calcium signaling in T lymphocytes was detected by flow cytometry and microscopy (fluorescence and confocal). In the presence of antibody, the percentage of responding T cells was reduced from 58.14% to 34.29%. As expected, anti-CD83 antibodies also reduced the proliferation of T lymphocytes (as assessed by CFSE dilution). Finally, in the absence of extracellular calcium, CD83 antibodies abrogated T cell signaling induced by allogeneic mDCs, suggesting that the presence of CD83 in mDC membranes enhances T lymphocyte proliferation by boosting calcium release from intracellular stores in these cells.
Collapse
Affiliation(s)
- Mariana Pereira Pinho
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, Brazil
| | - Isabella Katz Migliori
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, Brazil
| | | | | |
Collapse
|
26
|
Guo Y, Li R, Song X, Zhong Y, Wang C, Jia H, Wu L, Wang D, Fang F, Ma J, Kang W, Sun J, Tian Z, Xiao W. The expression and characterization of functionally active soluble CD83 by Pichia pastoris using high-density fermentation. PLoS One 2014; 9:e89264. [PMID: 24586642 PMCID: PMC3930729 DOI: 10.1371/journal.pone.0089264] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2013] [Accepted: 01/17/2014] [Indexed: 11/18/2022] Open
Abstract
CD83 is a highly glycosylated type I transmembrane glycoprotein that belongs to the immunoglobulin superfamily. CD83 is upregulated during dendritic cell (DC) maturation, which is critical for the initiation of adaptive immune responses. The soluble isoform of CD83 (sCD83) is encoded by alternative splicing from full-length CD83 mRNA and inhibits DC maturation, which suggests that sCD83 acts as a potential immune suppressor. In this study, we developed a sound strategy to express functional sCD83 from Pichia pastoris in extremely high-density fermentation. Purified sCD83 was expressed as a monomer at a yield of more than 200 mg/L and contained N-linked glycosylation sites that were characterized by PNGase F digestion. In vitro tests indicated that recombinant sCD83 bound to its putative counterpart on monocytes and specifically blocked the binding of anti-CD83 antibodies to cell surface CD83 on DCs. Moreover, sCD83 from yeast significantly suppressed ConA-stimulated PBMC proliferation. Therefore, sCD83 that was expressed from the P. pastoris was functionally active and may be used for in vivo and in vitro studies as well as future clinical applications.
Collapse
Affiliation(s)
- Yugang Guo
- Hefei National Laboratory for Physical Sciences at Microscale, Innovation Center for Cell Biology, School of Life Sciences, University of Science and Technology of China, Hefei, China
- Center of Medical Biotechnology of Anhui Province, University of Science and Technology of China, Hefei, China
| | - Rui Li
- Hefei National Laboratory for Physical Sciences at Microscale, Innovation Center for Cell Biology, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Xiaoping Song
- Department of Pharmacy, Anhui Medical College, Hefei, China
| | - Yongjun Zhong
- Hefei National Laboratory for Physical Sciences at Microscale, Innovation Center for Cell Biology, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Chenguang Wang
- Hefei National Laboratory for Physical Sciences at Microscale, Innovation Center for Cell Biology, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Hao Jia
- Hefei National Laboratory for Physical Sciences at Microscale, Innovation Center for Cell Biology, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Lidan Wu
- Hefei National Laboratory for Physical Sciences at Microscale, Innovation Center for Cell Biology, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Dong Wang
- Hefei National Laboratory for Physical Sciences at Microscale, Innovation Center for Cell Biology, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Fang Fang
- Hefei National Laboratory for Physical Sciences at Microscale, Innovation Center for Cell Biology, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Jiajia Ma
- Center of Medical Biotechnology of Anhui Province, University of Science and Technology of China, Hefei, China
| | - Wenyao Kang
- Hefei National Laboratory for Physical Sciences at Microscale, Innovation Center for Cell Biology, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Jie Sun
- Hefei National Laboratory for Physical Sciences at Microscale, Innovation Center for Cell Biology, School of Life Sciences, University of Science and Technology of China, Hefei, China
- Center of Medical Biotechnology of Anhui Province, University of Science and Technology of China, Hefei, China
| | - Zhigang Tian
- Hefei National Laboratory for Physical Sciences at Microscale, Innovation Center for Cell Biology, School of Life Sciences, University of Science and Technology of China, Hefei, China
- Center of Medical Biotechnology of Anhui Province, University of Science and Technology of China, Hefei, China
| | - Weihua Xiao
- Hefei National Laboratory for Physical Sciences at Microscale, Innovation Center for Cell Biology, School of Life Sciences, University of Science and Technology of China, Hefei, China
- Center of Medical Biotechnology of Anhui Province, University of Science and Technology of China, Hefei, China
| |
Collapse
|
27
|
Yuan Y, Wan L, Chen Y, Shi M, Wang C, Zhao J, Lu X, Wang H, Lu Y, Cheng J. Production and characterization of human soluble CD83 fused with the fragment crystallizable region of human IgG1 in Pichia pastoris. Appl Microbiol Biotechnol 2013; 97:9409-17. [PMID: 23392767 DOI: 10.1007/s00253-013-4732-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Revised: 01/16/2013] [Accepted: 01/19/2013] [Indexed: 02/05/2023]
Abstract
The cell surface protein CD83 belongs to the immunoglobulin superfamily and is highly expressed on mature dendritic cells. The soluble form of CD83, sCD83, is a potential immune suppressor. In a previous study, recombinant soluble CD83 was expressed in Escherichia coli, resulting in a lack of functional glycosylation. Although eukaryotic cell systems for producing sCD83 offer the advantages of protein processing, folding, and posttranslational modification, these systems are complicated, expensive, and produce low levels of protein. To obtain more efficient expression of sCD83, we expressed human sCD83 fused with fragment crystallizable region of human IgG1 (hIgG1 Fc) in Pichia pastoris. Under the optimal conditions (time of induction, 48 h; inoculum density (OD600), 80; concentration of methanol, 3.0 %; pH 7.0-8.0; concentration of casamino acid, 5.0 %), the purified human sCD83-hIgG1 Fc (hsCD83-Ig) fusion protein existed as dimers at 25-30 mg/L culture. Treatment with PNGase F showed that purified hsCD83-Ig was modified by N-linked glycosylation. Moreover, the hsCD83-Ig expressed in the P. pastoris system could suppress lymphocyte proliferation in ConA-stimulated and one-way mixed lymphocyte reaction systems. Thus, hsCD83-Ig expressed in P. pastoris is functional and may be used in experimental therapies for graft rejection, graft-versus-host disease, and autoimmune diseases.
Collapse
Affiliation(s)
- Yujia Yuan
- Key Laboratory of Transplant Engineering and Immunology, Ministry of Health, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Starke C, Steinkasserer A, Voll RE, Zinser E. Soluble human CD83 ameliorates lupus in NZB/W F1 mice. Immunobiology 2013; 218:1411-5. [PMID: 23886695 DOI: 10.1016/j.imbio.2013.06.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Revised: 05/23/2013] [Accepted: 06/05/2013] [Indexed: 11/28/2022]
Abstract
In the present study we explored the immunomodulatory potential of prokaryotically expressed soluble CD83 in the treatment of murine lupus using the NZB/W F1 mouse model. Therefore female NZB/W F1 lupus mice were treated either with sCD83 or PBS for 4 weeks. sCD83 treated mice showed a significantly delayed onset of anti-dsDNA autoantibody production when compared with the control group. Importantly, during the treatment period with sCD83 none of the mice showed elevated levels of anti-dsDNA autoantibodies. In addition, NZB/W F1 mice which received sCD83 displayed lower concentrations of anti-histone IgG autoantibodies. Furthermore, there was no difference in total IgG antibodies, indicating a modulatory role for sCD83 in the production of self-reactive antibodies without decreasing total IgG. These results indicate that administration of sCD83 has profound immune-modulatory effects on the induction of autoantibodies in NZB/W F1 lupus mice and may thus be a promising approach to interfere with autoimmunity in SLE and other autoantibody-driven diseases.
Collapse
Affiliation(s)
- Charlotte Starke
- Department of Internal Medicine 3 and Institute of Clinical Immunology, Nikolaus-Fiebiger Center, University of Erlangen-Nuremberg, Erlangen, Germany; Department of Internal Medicine 3, University of Technology, Dresden, Germany
| | | | | | | |
Collapse
|
29
|
Bock F, Rössner S, Onderka J, Lechmann M, Pallotta MT, Fallarino F, Boon L, Nicolette C, DeBenedette MA, Tcherepanova IY, Grohmann U, Steinkasserer A, Cursiefen C, Zinser E. Topical application of soluble CD83 induces IDO-mediated immune modulation, increases Foxp3+ T cells, and prolongs allogeneic corneal graft survival. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2013; 191:1965-75. [PMID: 23851696 DOI: 10.4049/jimmunol.1201531] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Modulation of immune responses is one of the main research aims in transplant immunology. In this study, we investigate the local immunomodulatory properties of soluble CD83 (sCD83) at the graft-host interface using the high-risk corneal transplantation model. In this model, which mimics the inflammatory status and the preexisting vascularization of high-risk patients undergoing corneal transplantation, allogeneic donor corneas are transplanted onto sCD83-treated recipient animals. This model allows the direct and precise application of the immune modulator at the transplantation side. Interestingly, sCD83 was able to prolong graft survival after systemic application as well as after topical application, which is therapeutically more relevant. The therapeutic effect was accompanied by an increase in the frequency of regulatory T cells and was mediated by the immune-regulatory enzyme IDO and TGF-β. In vitro, sCD83 induced long-term IDO expression in both conventional and plasmacytoid dendritic cells via autocrine or paracrine production of TGF-β, a cytokine previously shown to be an essential mediator of IDO-dependent, long-term tolerance. These findings open new treatment avenues for local immune modulation after organ and tissue transplantation.
Collapse
MESH Headings
- Administration, Ophthalmic
- Allografts
- Animals
- Antigens, CD/administration & dosage
- Antigens, CD/immunology
- Antigens, CD/therapeutic use
- Bone Marrow Cells/immunology
- Cells, Cultured
- Coculture Techniques
- Corneal Transplantation
- Dendritic Cells/immunology
- Drug Evaluation, Preclinical
- Enzyme Induction/drug effects
- Female
- Forkhead Transcription Factors/analysis
- Graft Enhancement, Immunologic
- Graft Survival
- Immunoglobulins/administration & dosage
- Immunoglobulins/immunology
- Immunoglobulins/therapeutic use
- Immunologic Factors/administration & dosage
- Immunologic Factors/immunology
- Immunologic Factors/therapeutic use
- Indoleamine-Pyrrole 2,3,-Dioxygenase/biosynthesis
- Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics
- Indoleamine-Pyrrole 2,3,-Dioxygenase/physiology
- Injections, Intraperitoneal
- Membrane Glycoproteins/administration & dosage
- Membrane Glycoproteins/immunology
- Membrane Glycoproteins/therapeutic use
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Premedication
- Recombinant Proteins/administration & dosage
- Recombinant Proteins/immunology
- Recombinant Proteins/therapeutic use
- Solubility
- T-Lymphocyte Subsets/immunology
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- Transforming Growth Factor beta/administration & dosage
- Transforming Growth Factor beta/physiology
- Transforming Growth Factor beta/therapeutic use
- Transplantation Tolerance/drug effects
- CD83 Antigen
Collapse
Affiliation(s)
- Felix Bock
- Department of Ophthalmology, University of Cologne, 50931 Cologne, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Stein MF, Lang S, Winkler TH, Deinzer A, Erber S, Nettelbeck DM, Naschberger E, Jochmann R, Stürzl M, Slany RK, Werner T, Steinkasserer A, Knippertz I. Multiple interferon regulatory factor and NF-κB sites cooperate in mediating cell-type- and maturation-specific activation of the human CD83 promoter in dendritic cells. Mol Cell Biol 2013; 33:1331-44. [PMID: 23339870 PMCID: PMC3624272 DOI: 10.1128/mcb.01051-12] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Accepted: 01/14/2013] [Indexed: 02/08/2023] Open
Abstract
CD83 is one of the best-known surface markers for fully mature dendritic cells (mature DCs), and its cell-type- and maturation-specific regulation makes the CD83 promoter an interesting tool for the genetic modulation of DCs. To determine the mechanisms regulating this DC- and maturation-specific CD83 expression, chromatin immunoprecipitation (ChIP)-on-chip microarray, biocomputational, reporter, electrophoretic mobility shift assay (EMSA), and ChIP analyses were performed. These studies led to the identification of a ternary transcriptional activation complex composed of an upstream regulatory element, a minimal promoter, and an enhancer, which have not been reported in this arrangement for any other gene so far. Notably, these DNA regions contain a complex framework of interferon regulatory factor (IRF)- and NF-κB transcription factor-binding sites mediating their arrangement. Mutation of any of the IRF-binding sites resulted in a significant loss of promoter activity, whereas overexpression of NF-κB transcription factors clearly enhanced transcription. We identified IRF-1, IRF-2, IRF-5, p50, p65, and cRel to be involved in regulating maturation-specific CD83 expression in DCs. Therefore, the characterization of this promoter complex not only contributes to the knowledge of DC-specific gene regulation but also suggests the involvement of a transcriptional module with binding sites separated into distinct regions in transcriptional activation as well as cell-type- and maturation-specific transcriptional targeting of DCs.
Collapse
Affiliation(s)
- Marcello F. Stein
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | - Stefan Lang
- Department of Biology, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Thomas H. Winkler
- Department of Biology, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Andrea Deinzer
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | - Sebastian Erber
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | - Dirk M. Nettelbeck
- Helmholtz University Group Oncolytic Adenoviruses at the DKFZ (German Cancer Research Center) and Department of Dermatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Elisabeth Naschberger
- Division of Molecular and Experimental Surgery, Department of Surgery, University Medical Center Erlangen, Erlangen, Germany
| | - Ramona Jochmann
- Division of Molecular and Experimental Surgery, Department of Surgery, University Medical Center Erlangen, Erlangen, Germany
| | - Michael Stürzl
- Division of Molecular and Experimental Surgery, Department of Surgery, University Medical Center Erlangen, Erlangen, Germany
| | - Robert K. Slany
- Department of Genetics, University Erlangen, Erlangen, Germany
| | - Thomas Werner
- Genomatix Software GmbH, Munich, Germany
- Internal Medicine, Nephrology, University of Michigan, Ann Arbor, Michigan, USA
| | - Alexander Steinkasserer
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | - Ilka Knippertz
- Department of Immune Modulation at the Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| |
Collapse
|
31
|
Ehlers C, Schirmer S, Kehlenbach RH, Hauber J, Chemnitz J. Post-transcriptional regulation of CD83 expression by AUF1 proteins. Nucleic Acids Res 2013; 41:206-19. [PMID: 23161671 PMCID: PMC3592417 DOI: 10.1093/nar/gks1069] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Revised: 09/27/2012] [Accepted: 10/11/2012] [Indexed: 12/31/2022] Open
Abstract
Mature dendritic cells (DC), activated lymphocytes, mononuclear cells and neutrophils express CD83, a surface protein apparently necessary for effective DC-mediated activation of naïve T-cells and T-helper cells, thymic T-cell maturation and the regulation of B-cell activation and homeostasis. Although a defined ligand of CD83 remains elusive, the multiple cellular subsets expressing CD83, as well as its numerous potential implications in immunological processes suggest that CD83 plays an important regulatory role in the mammalian immune system. Lately, nucleocytoplasmic translocation of CD83 mRNA was shown to be mediated by direct interaction between the shuttle protein HuR and a novel post-transcriptional regulatory element (PRE) located in the CD83 transcript's coding region. Interestingly, this interaction commits the CD83 mRNA to efficient nuclear export through the CRM1 protein translocation pathway. More recently, the cellular phosphoprotein and HuR ligand ANP32B (APRIL) was demonstrated to be directly involved in this intracellular transport process by linking the CD83 mRNA:HuR ribonucleoprotein (RNP) complex with the CRM1 export receptor. Casein kinase II regulates this process by phosphorylating ANP32B. Here, we identify another RNA binding protein, AUF1 (hnRNP D) that directly interacts with CD83 PRE. Unlike HuR:PRE binding, this interaction has no impact on intracellular trafficking of CD83 mRNA-containing complexes; but it does regulate translation of CD83 mRNA. Thus, our data shed more light on the complex process of post-transcriptional regulation of CD83 expression. Interfering with this process may provide a novel strategy for inhibiting CD83, and thereby cellular immune activation.
Collapse
Affiliation(s)
- Christina Ehlers
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Martinistrasse 52, D-20251 Hamburg and Zentrum für Biochemie und Molekulare Zellbiologie, Universität Göttingen, Humboldtallee 23, D-37073 Göttingen, Germany
| | - Susann Schirmer
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Martinistrasse 52, D-20251 Hamburg and Zentrum für Biochemie und Molekulare Zellbiologie, Universität Göttingen, Humboldtallee 23, D-37073 Göttingen, Germany
| | - Ralph H. Kehlenbach
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Martinistrasse 52, D-20251 Hamburg and Zentrum für Biochemie und Molekulare Zellbiologie, Universität Göttingen, Humboldtallee 23, D-37073 Göttingen, Germany
| | - Joachim Hauber
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Martinistrasse 52, D-20251 Hamburg and Zentrum für Biochemie und Molekulare Zellbiologie, Universität Göttingen, Humboldtallee 23, D-37073 Göttingen, Germany
| | - Jan Chemnitz
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Martinistrasse 52, D-20251 Hamburg and Zentrum für Biochemie und Molekulare Zellbiologie, Universität Göttingen, Humboldtallee 23, D-37073 Göttingen, Germany
| |
Collapse
|
32
|
Jonker M, Wubben J, Haanstra K, Vierboom M, 't Hart B. Comparative analysis of inflammatory infiltrates in collagen-induced arthritis, kidney graft rejection and delayed-type hypersensitivity in non-human primates. Inflamm Res 2012; 62:181-94. [PMID: 23064655 DOI: 10.1007/s00011-012-0564-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Revised: 08/15/2012] [Accepted: 10/03/2012] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVES Non-human primates are immunologically closely related to humans providing relevant models of inflammatory disorders often used to evaluate new immunomodulating therapies. The aim of the study was to compare inflammatory infiltrates of acute graft rejection (AR) and collagen-induced arthritis (CIA) to delayed-type hypersensitivity (DTH) reactions as the latter model may serve as a less invasive animal model. MATERIALS AND METHODS Tissue samples of AR, CIA and DTH were obtained from rhesus monkeys used in several pre-clinical studies. The infiltrate composition was determined by immunohistochemical analysis. RESULTS The infiltrates in AR consisted of T cells, macrophages and B cells. The presence of lymphoid structures in AR suggested ongoing intragraft immune activation. The synovia of CIA contained predominantly macrophages and few T cells. The DTH infiltrates were dominated by T cells when the challenged was ovalbumin (OVA) and by macrophages when the challenge was tetanus toxoid (TT). CONCLUSIONS The histology of AR resembles aspects of DTH to OVA while that of CIA showed similarities of the DTH to TT. The DTH reaction could serve as a model to study immunomodulating drugs for acute rejection and the acute inflammatory phase of autoimmunity.
Collapse
Affiliation(s)
- Margreet Jonker
- Biomedical Primate Research Centre, PO BOX 3306, 2280 GH Rijswijk, Netherlands.
| | | | | | | | | |
Collapse
|
33
|
Pőcze B, Németh P, Langer R. [Recent options in drug therapy after solid organ transplantation]. Orv Hetil 2012; 153:1294-301. [PMID: 22890176 DOI: 10.1556/oh.2012.29343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Solid organ transplantation has shown improvement in patient and graft survival rates due to the development of immunosuppression in the last fifty years; however only the last two decades led to the development of new, baseline immunosuppressive drugs that avoid the unlikely side effects of calcineurin inhibitors, especially nephrotoxicity. The transplanted organ is foreign to the host and, therefore, it induces a complex immune response of the recipient. In this review, a brief outline of immune response is given, followed by the introduction of new immunosuppressive drugs acting via variant pathways. These are compounds which are already in use or becoming shortly available and are potential future alternatives for the calcineurin inhibitors. This paper highlights the role of co-stimulation blockade with belatacept and the recently even more intensively studied field of tolerance induction.
Collapse
Affiliation(s)
- Balázs Pőcze
- Semmelweis Egyetem, Általános Orvostudományi Kar Transzplantációs és Sebészeti Klinika Budapest.
| | | | | |
Collapse
|
34
|
CD11c controls herpes simplex virus 1 responses to limit virus replication during primary infection. J Virol 2011; 85:9945-55. [PMID: 21775452 DOI: 10.1128/jvi.05208-11] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
CD11c is expressed on the surface of dendritic cells (DCs) and is one of the main markers for identification of DCs. DCs are the effectors of central innate immune responses, but they also affect acquired immune responses to infection. However, how DCs influence the efficacy of adaptive immunity is poorly understood. Here, we show that CD11c(+) DCs negatively orchestrate both adaptive and innate immunity against herpes simplex virus type 1 (HSV-1) ocular infection. The effectiveness and quantity of virus-specific CD8(+) T cell responses are increased in CD11c-deficient animals. In addition, the levels of CD83, CD11b, alpha interferon (IFN-α), and IFN-β, but not IFN-γ, were significantly increased in CD11c-deficient animals. Higher levels of IFN-α, IFN-β, and CD8(+) T cells in the CD11c-deficient mice may have contributed to lower virus replication in the eye and trigeminal ganglia (TG) during the early period of infection than in wild-type mice. However, the absence of CD11c did not influence survival, severity of eye disease, or latency. Our studies provide for the first time evidence that CD11c expression may abrogate the ability to reduce primary virus replication in the eye and TG via higher activities of type 1 interferon and CD8(+) T cell responses.
Collapse
|
35
|
Ezzelarab M, Thomson AW. Tolerogenic dendritic cells and their role in transplantation. Semin Immunol 2011; 23:252-63. [PMID: 21741270 DOI: 10.1016/j.smim.2011.06.007] [Citation(s) in RCA: 119] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Accepted: 06/10/2011] [Indexed: 01/09/2023]
Abstract
The pursuit of clinical transplant tolerance has led to enhanced understanding of mechanisms underlying immune regulation, including the characterization of immune regulatory cells, in particular antigen-presenting cells (APC) and regulatory T cells (Treg), that may play key roles in promoting operational tolerance. Dendritic cells (DC) are highly efficient APC that have been studied extensively in rodents and humans, and more recently in non-human primates. Owing to their ability to regulate both innate and adaptive immune responses, DC are considered to play crucial roles in directing the alloimmune response towards transplant tolerance or rejection. Mechanisms via which they can promote central and peripheral tolerance include clonal deletion, the induction of Treg, and inhibition of memory T cell responses. These properties have led to the use of tolerogenic DC as a therapeutic strategy to promote organ transplant tolerance. In rodents, infusion of donor- or recipient-derived tolerogenic DC can extensively prolong donor-specific allograft survival, in association with regulation of the host T cell response. In clinical transplantation, progress has been made in monitoring DC in relation to graft outcome, including studies in operational liver transplant tolerance. Although clinical trials involving immunotherapeutic DC for patients with cancer are ongoing, implementation of human DC therapy in clinical transplantation will require assessment of various critical issues. These include cell isolation and purification techniques, source, route and timing of administration, and combination immunosuppressive therapy. With ongoing non-human primate studies focused on DC therapy, these logistics can be investigated seeking the optimal approaches. The scientific rationale for implementation of tolerogenic DC therapy to promote clinical transplant tolerance is strong. Evaluation of technical and therapeutic logistic issues is an important next step prior to the application of tolerogenic DC in clinical organ transplantation.
Collapse
Affiliation(s)
- Mohamed Ezzelarab
- Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh School of Medicine, 200 Lothrop Street, BST W1540, Pittsburgh, PA 15261, USA
| | | |
Collapse
|