1
|
Bettonville M, d'Aria S, Weatherly K, Porporato PE, Zhang J, Bousbata S, Sonveaux P, Braun MY. Long-term antigen exposure irreversibly modifies metabolic requirements for T cell function. eLife 2018; 7:30938. [PMID: 29911570 PMCID: PMC6025959 DOI: 10.7554/elife.30938] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 06/14/2018] [Indexed: 12/11/2022] Open
Abstract
Energy metabolism is essential for T cell function. However, how persistent antigenic stimulation affects T cell metabolism is unknown. Here, we report that long-term in vivo antigenic exposure induced a specific deficit in numerous metabolic enzymes. Accordingly, T cells exhibited low basal glycolytic flux and limited respiratory capacity. Strikingly, blockade of inhibitory receptor PD-1 stimulated the production of IFNγ in chronic T cells, but failed to shift their metabolism towards aerobic glycolysis, as observed in effector T cells. Instead, chronic T cells appeared to rely on oxidative phosphorylation (OXPHOS) and fatty acid oxidation (FAO) to produce ATP for IFNγ synthesis. Check-point blockade, however, increased mitochondrial production of superoxide and reduced viability and effector function. Thus, in the absence of a glycolytic switch, PD-1-mediated inhibition appears essential for limiting oxidative metabolism linked to effector function in chronic T cells, thereby promoting survival and functional fitness.
Collapse
Affiliation(s)
- Marie Bettonville
- Institute for Medical Immunology, Université Libre de Bruxelles, Gosselies, Belgium
| | - Stefania d'Aria
- Institute for Medical Immunology, Université Libre de Bruxelles, Gosselies, Belgium
| | - Kathleen Weatherly
- Institute for Medical Immunology, Université Libre de Bruxelles, Gosselies, Belgium
| | - Paolo E Porporato
- Pole of Pharmacology & Therapeutics, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Jinyu Zhang
- Institute for Medical Immunology, Université Libre de Bruxelles, Gosselies, Belgium
| | - Sabrina Bousbata
- Laboratory of Molecular Parasitology, Proteomic Platform, Institute of Molecular Biology and Medicine, Université Libre de Bruxelles, Gosselies, Belgium
| | - Pierre Sonveaux
- Pole of Pharmacology & Therapeutics, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Michel Y Braun
- Institute for Medical Immunology, Université Libre de Bruxelles, Gosselies, Belgium
| |
Collapse
|
2
|
Schiechl G, Hermann FJ, Rodriguez Gomez M, Kutzi S, Schmidbauer K, Talke Y, Neumayer S, Goebel N, Renner K, Brühl H, Karasuyama H, Obata-Ninomiya K, Utpatel K, Evert M, Hirt SW, Geissler EK, Fichtner-Feigl S, Mack M. Basophils Trigger Fibroblast Activation in Cardiac Allograft Fibrosis Development. Am J Transplant 2016; 16:2574-88. [PMID: 26932231 DOI: 10.1111/ajt.13764] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 01/29/2016] [Accepted: 02/22/2016] [Indexed: 01/25/2023]
Abstract
Fibrosis is a major component of chronic cardiac allograft rejection. Although several cell types are able to produce collagen, resident (donor-derived) fibroblasts are mainly responsible for excessive production of extracellular matrix proteins. It is currently unclear which cells regulate production of connective tissue elements in allograft fibrosis and how basophils, as potential producers of profibrotic cytokines, are involved this process. We studied this question in a fully MHC-mismatched model of heart transplantation with transient depletion of CD4(+) T cells to largely prevent acute rejection. The model is characterized by myocardial infiltration of leukocytes and development of interstitial fibrosis and allograft vasculopathy. Using depletion of basophils, IL-4-deficient recipients and IL-4 receptor-deficient grafts, we showed that basophils and IL-4 play crucial roles in activation of fibroblasts and development of fibrotic organ remodeling. In the absence of CD4(+) T cells, basophils are the predominant source of IL-4 in the graft and contribute to expansion of myofibroblasts, interstitial deposition of collagen and development of allograft vasculopathy. Our results indicated that basophils trigger the production of various connective tissue elements by myofibroblasts. Basophil-derived IL-4 may be an attractive target for treatment of chronic allograft rejection.
Collapse
Affiliation(s)
- G Schiechl
- Department of Internal Medicine II, Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - F J Hermann
- Department of Internal Medicine II, Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - M Rodriguez Gomez
- Department of Internal Medicine II, Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - S Kutzi
- Department of Internal Medicine II, Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - K Schmidbauer
- Department of Internal Medicine II, Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Y Talke
- Department of Internal Medicine II, Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - S Neumayer
- Department of Internal Medicine II, Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - N Goebel
- Department of Internal Medicine II, Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - K Renner
- Department of Internal Medicine II, Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - H Brühl
- Department of Internal Medicine I, University Hospital Regensburg, Regensburg, Germany
| | - H Karasuyama
- Department of Immune Regulation, Tokyo Medical and Dental University Graduate School, Tokyo, Japan
| | - K Obata-Ninomiya
- Department of Immune Regulation, Tokyo Medical and Dental University Graduate School, Tokyo, Japan
| | - K Utpatel
- Department of Pathology, University Hospital Regensburg, Regensburg, Germany
| | - M Evert
- Department of Pathology, University Hospital Regensburg, Regensburg, Germany
| | - S W Hirt
- Department of Cardiothoracic Surgery, University Hospital Regensburg, Regensburg, Germany
| | - E K Geissler
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - S Fichtner-Feigl
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany.,RCI Regensburg Center for Interventional Immunology, University of Regensburg, Regensburg, Germany
| | - M Mack
- Department of Internal Medicine II, Nephrology, University Hospital Regensburg, Regensburg, Germany.,RCI Regensburg Center for Interventional Immunology, University of Regensburg, Regensburg, Germany
| |
Collapse
|
3
|
Xie J, Wang Y, Bao J, Ma Y, Zou Z, Tang Z, Dong R, Wen H. Immune tolerance induced by RelB short-hairpin RNA interference dendritic cells in liver transplantation. J Surg Res 2013. [DOI: 10.1016/j.jss.2012.10.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
4
|
Li D, Tsang JYS, Peng J, Ho DHH, Chan YK, Zhu J, Lui VCH, Xu A, Lamb JR, Tam PKH, Chen Y. Adiponectin mediated MHC class II mismatched cardiac graft rejection in mice is IL-4 dependent. PLoS One 2012; 7:e48893. [PMID: 23155424 PMCID: PMC3498365 DOI: 10.1371/journal.pone.0048893] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Accepted: 10/03/2012] [Indexed: 11/18/2022] Open
Abstract
Background Adiponectin regulates glucose and fatty-acid metabolism but its role in chronic graft rejection mediated by Th2 cytokines remains ill-defined. Methodology/Principal Findings Wild type and adiponectin-null mice were used as graft recipients in mouse MHC class II disparate cardiac transplantation (bm12 toB6) and the graft rejection was monitored. In adiponectin-null mice we observed that the cellular infiltrate of eosinophils, CD4+ and CD8+ T cells was reduced in grafts compared to the controls as was collagen deposition and vessel occlusion. A similar outcome was observed for skin transplants except that neutrophil infiltration was increased. Low levels of IL-4 were detected in the grafts and serum. The effect of adiponectin signaling on IL-4 expression was further investigated. Treatment with AMPK and p38 MAPK inhibitors blocked adiponectin enhanced T cell proliferation in mixed lymphocyte reactions. Inhibition of AMPK reduced eosinophil infiltration in skin grafts in wild type recipients and in contrast AMPK activation increased eosinophils in adiponectin-null recipients. The addition of adiponectin increased IL-4 production by the T cell line EL4 with augmented nuclear GATA-3 and phospho-STAT6 expression which were suppressed by knockdown of adiponectin receptor 1 and 2. Conclusions Our results demonstrate a direct effect of adiponectin on IL-4 expression which contributes to Th2 cytokine mediated rejection in mouse MHC class II histoincompatible transplants. These results add to our understanding of the interrelationship of metabolism and immune regulation and raise the possibility that AMPK inhibitors may be beneficial in selected types of rejection.
Collapse
Affiliation(s)
- Daxu Li
- Paediatric Surgery Division, Department of Surgery, The University of Hong Kong, Hong Kong SAR, China
| | - Julia Y. S. Tsang
- Paediatric Surgery Division, Department of Surgery, The University of Hong Kong, Hong Kong SAR, China
| | - Jiao Peng
- Paediatric Surgery Division, Department of Surgery, The University of Hong Kong, Hong Kong SAR, China
| | - Derek H. H. Ho
- Department of Chemistry, The University of Hong Kong, Hong Kong SAR, China
| | - Yee Kwan Chan
- School of Biological Science, The University of Hong Kong, Hong Kong SAR, China
| | - Jiang Zhu
- Paediatric Surgery Division, Department of Surgery, The University of Hong Kong, Hong Kong SAR, China
| | - Vincent C. H. Lui
- Paediatric Surgery Division, Department of Surgery, The University of Hong Kong, Hong Kong SAR, China
| | - Aimin Xu
- Department of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Jonathan R. Lamb
- Division of Cell and Molecular Biology, Faculty of Natural Sciences, Imperial College London, London, United Kingdom
| | - Paul K. H. Tam
- Paediatric Surgery Division, Department of Surgery, The University of Hong Kong, Hong Kong SAR, China
- * E-mail: (YC); (PT)
| | - Yan Chen
- Paediatric Surgery Division, Department of Surgery, The University of Hong Kong, Hong Kong SAR, China
- Department of Chemistry, The University of Hong Kong, Hong Kong SAR, China
- * E-mail: (YC); (PT)
| |
Collapse
|
5
|
Tolerogenic dendritic cells generated by RelB silencing using shRNA prevent acute rejection. Cell Immunol 2012; 274:12-8. [PMID: 22464914 DOI: 10.1016/j.cellimm.2012.02.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2011] [Revised: 01/27/2012] [Accepted: 02/29/2012] [Indexed: 12/21/2022]
Abstract
It is well known that adoptive transfer of donor-derived tolerogenic dendritic cells (DCs) helps to induce immune tolerance. RelB, one of NF-κB subunits, is a critical element involved in DC maturation. In the present study, our results showed tolerogenic DCs could be acquired via silencing RelB using small interfering RNA. Compared with imDCs, the tolerogenic DCs had more potent ability to inhibit mixed lymphocyte reaction (MLR) and down-regulate Th1 cytokines and prompt the production of Th2 cytokines. They both mediated immune tolerance via the increased of T cell apoptosis and generation of regulatory T cells. Administration of donor-derived tolerogenic DCs significantly prevented the allograft rejection and prolonged the survival time in a murine heart transplantation model. Our results demonstrate donor-derived, RelB-shRNA induced tolerogenic DCs can significantly induce immune tolerance in vitro and in vivo.
Collapse
|
6
|
Shimizu K, Libby P, Rocha VZ, Folco EJ, Shubiki R, Grabie N, Jang S, Lichtman AH, Shimizu A, Hogg N, Simon DI, Mitchell RN, Croce K. Loss of myeloid related protein-8/14 exacerbates cardiac allograft rejection. Circulation 2011; 124:2920-32. [PMID: 22144572 PMCID: PMC3277828 DOI: 10.1161/circulationaha.110.009910] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2010] [Accepted: 10/26/2011] [Indexed: 02/03/2023]
Abstract
BACKGROUND The calcium-binding proteins myeloid-related protein (MRP)-8 (S100A8) and MRP-14 (S100A9) form MRP-8/14 heterodimers (S100A8/A9, calprotectin) that regulate myeloid cell function and inflammatory responses and serve as early serum markers for monitoring acute allograft rejection. Despite functioning as a proinflammatory mediator, the pathophysiological role of MRP-8/14 complexes in cardiovascular disease is incompletely defined. This study investigated the role of MRP-8/14 in cardiac allograft rejection using MRP-14(-/-) mice that lack MRP-8/14 complexes. METHODS AND RESULTS We examined parenchymal rejection after major histocompatibility complex class II allomismatched cardiac transplantation (bm12 donor heart and B6 recipients) in wild-type (WT) and MRP-14(-/-) recipients. Allograft survival averaged 5.9±2.9 weeks (n=10) in MRP-14(-/-) recipients compared with >12 weeks (n=15; P<0.0001) in WT recipients. Two weeks after transplantation, allografts in MRP-14(-/-) recipients had significantly higher parenchymal rejection scores (2.8±0.8; n=8) than did WT recipients (0.8±0.8; n=12; P<0.0001). Compared with WT recipients, allografts in MRP-14(-/-) recipients had significantly increased T-cell and macrophage infiltration and increased mRNA levels of interferon-γ and interferon-γ-associated chemokines (CXCL9, CXCL10, and CXCL11), interleukin-6, and interleukin-17 with significantly higher levels of Th17 cells. MRP-14(-/-) recipients also had significantly more lymphocytes in the adjacent para-aortic lymph nodes than did WT recipients (cells per lymph node: 23.7±0.7×10(5) for MRP-14(-/-) versus 6.0±0.2×10(5) for WT; P<0.0001). The dendritic cells (DCs) of the MRP-14(-/-) recipients of bm12 hearts expressed significantly higher levels of the costimulatory molecules CD80 and CD86 than did those of WT recipients 2 weeks after transplantation. Mixed leukocyte reactions with allo-endothelial cell-primed MRP-14(-/-) DCs resulted in significantly higher antigen-presenting function than reactions using WT DCs. Ovalbumin-primed MRP-14(-/-) DCs augmented proliferation of OT-II (ovalbumin-specific T cell receptor transgenic) CD4(+) T cells with increased interleukin-2 and interferon-γ production. Cardiac allografts of B6 major histocompatibility complex class II(-/-) hosts and of B6 WT hosts receiving MRP-14(-/-) DCs had significantly augmented inflammatory cell infiltration and accelerated allograft rejection compared with WT DCs from transferred recipient allografts. Bone marrow-derived MRP-14(-/-) DCs infected with MRP-8 and MRP-14 retroviral vectors showed significantly decreased CD80 and CD86 expression compared with controls, indicating that MRP-8/14 regulates B7-costimulatory molecule expression. CONCLUSIONS Our results indicate that MRP-14 regulates B7 molecule expression and reduces antigen presentation by DCs and subsequent T-cell priming. The absence of MRP-14 markedly increased T-cell activation and exacerbated allograft rejection, indicating a previously unrecognized role for MRP-14 in immune cell biology.
Collapse
Affiliation(s)
- Koichi Shimizu
- Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, NRB7, Boston, MA 02115, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Cobbold SP, Adams E, Nolan KF, Regateiro FS, Waldmann H. Connecting the mechanisms of T-cell regulation: dendritic cells as the missing link. Immunol Rev 2010; 236:203-18. [PMID: 20636819 DOI: 10.1111/j.1600-065x.2010.00913.x] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A variety of different molecular mechanisms have been proposed to explain the suppressive action of regulatory T cells, including the production of anti-inflammatory cytokines, negative costimulatory ligands, indoleamine 2,3-dioxygenase-mediated tryptophan catabolism, CD73-mediated adenosine generation, and downregulation of antigen-presenting cells. Until now it has been unclear how important each of these different mechanisms might be and how they are coordinated. In this review, we examine the hypothesis that it is the interaction between regulatory T cells and dendritic cells that creates a local microenvironment depleted of essential amino acids and rich in adenosine that leads to the amplification of a range of different tolerogenic signals. These signals are all eventually integrated by mammalian target of rapamycin inhibition, which enables the induction of new forkhead box protein 3-expressing Tregs. If correct, this provides a molecular explanation for the in vivo phenomena of linked suppression and infectious tolerance.
Collapse
Affiliation(s)
- Stephen P Cobbold
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK.
| | | | | | | | | |
Collapse
|
8
|
Suppression of ongoing experimental autoimmune myasthenia gravis by transfer of RelB-silenced bone marrow dentritic cells is associated with a change from a T helper Th17/Th1 to a Th2 and FoxP3+ regulatory T-cell profile. Inflamm Res 2009; 59:197-205. [PMID: 19768385 DOI: 10.1007/s00011-009-0087-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2009] [Revised: 07/15/2009] [Accepted: 08/19/2009] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE To observe the therapeutic effect of RelB-silenced dendritic cells (DCs) in experimental autoimmune myasthenia gravis (EAMG), and further to investigate the mechanism of this immune system therapeutic. METHODS (1) RelB-silenced DCs and control DCs were prepared and the supernatants were collected for IL-12p70, IL-6, and IL-23 measurement by ELISA. (2) RelB-silenced DCs and control DCs were co-cultured with AChR-specific T cells, and the supernatant was collected to observe the IL-17, IFN-gamma, IL-4 production. (3) EAMG mice with clinical scores of 1 to 2 were collected and enrolled randomly into the RelB-silenced DC group or the control DC group. RelB-silenced DCs or an equal amount of control DCs were injected intravenously on days 0, 7, and 14 after enrollment. Clinical scores were evaluated every other day. Twenty days after allotment, serum from individual mice was collected to detect serum concentrations of anti-mouse AChR IgG, IgG1, IgG2b, and IgG2c. The splenocytes were isolated for analysis of lymphocyte proliferative responses, cytokine (IL-17, IFN-gamma, IL-4) production, and protein levels of RORgammat, T-bet, GATA-3, and FoxP3 (the special transcription factors of Th17, Th1, Th2, and Treg, respectively). RESULTS (1) RelB-silenced DCs produced significantly reduced amounts of IL-12p70, IL-6, and IL-23, as compared with control DCs. (2) RelB-silenced DCs spurred on the CD4(+) T cells from Th1/Th17 to the Th2 cell subset in the co-culture system. (3) Treatment with RelB-silenced DCs effectively reduced myasthenic symptoms and levels of serum anti-acetylcholine receptor autoantibody in mice with ongoing EAMG. Th17-related markers (RORgammat, IL-17) and Th1-related markers (T-bet, IFN-gamma) were downregulated, whereas Th2 markers (IL-4 and GATA3) and Treg marker (FoxP3) were upregulated. CONCLUSIONS RelB-silenced DCs were able to create a particular cytokine environment that was absent of inflammatory cytokines. RelB-silenced DCs provide a practical means to normalize the differentiation of the four T-cell subsets (Th17, Th1, Th2, and Treg) in vivo, and thus possess therapeutic potential in Th1/Th17-dominant autoimmune disorders such as myasthenia gravis.
Collapse
|
9
|
Bouazzaoui A, Spacenko E, Mueller G, Miklos S, Huber E, Holler E, Andreesen R, Hildebrandt GC. Chemokine and chemokine receptor expression analysis in target organs of acute graft-versus-host disease. Genes Immun 2009; 10:687-701. [PMID: 19571824 DOI: 10.1038/gene.2009.49] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
10
|
Abstract
PURPOSE OF REVIEW Due to the dominance of Th1 cytokines in rejection and the ability of Th2 cytokines, particularly IL-4, to inhibit Th1 responses, it has long been held that Th2 cytokines can improve transplant outcomes. Although there is some support for this, there is mounting evidence that IL-4 and Th2 cytokines can promote graft dysfunction. These disparate effects are reviewed. RECENT FINDINGS The role of Th2 cytokines in graft dysfunction is not necessarily due to promotion of humoral immunity, but is due to their ability to drive T-cell and non-T-cell responses including alternative activation of macrophages. Alternatively, activated macrophages compete with classically activated macrophages for arginine and they are mutually exclusive, analogous to mutual competition between Th1 and Th2 cells. Recent findings also point to two subsets of regulatory T cells (Tregs), each dependent on either Th1 or Th2 cytokines. In addition to its effects on bone marrow-derived cells, IL-4 affects parenchymal cells by signalling through the type II receptor, which consists of the IL-4R alpha chain (IL-4Ralpha) and the IL-13Ralpha1, which also binds IL-13. SUMMARY The effects of Th2 cytokines in transplantation depend on their cellular targets, the timing and form of administration and on Th2 cytokine-dependent Tregs.
Collapse
|
11
|
Incorporation of CD40 ligand into the envelope of pseudotyped single-cycle Simian immunodeficiency viruses enhances immunogenicity. J Virol 2008; 83:1216-27. [PMID: 19036823 DOI: 10.1128/jvi.01870-08] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
A vaccine for the prevention of human immunodeficiency virus (HIV) infection is desperately needed to control the AIDS pandemic. To address this problem, we developed vesicular stomatitis virus glycoprotein-pseudotyped replication-defective simian immunodeficiency viruses (dSIVs) as an AIDS vaccine strategy. The dSIVs retain characteristics of a live attenuated virus without the drawbacks of potential virulence caused by replicating virus. To improve vaccine immunogenicity, we incorporated CD40 ligand (CD40L) into the dSIV envelope. CD40L is one of the most potent stimuli for dendritic cell (DC) maturation and activation. Binding of CD40L to its receptor upregulates expression of major histocompatibility complex class I, class II, and costimulatory molecules on DCs and increases production of proinflammatory cytokines and chemokines, especially interleukin 12 (IL-12). This cytokine polarizes CD4(+) T cells to Th1-type immune responses. DC activation and mixed lymphocyte reaction (MLR) studies were performed to evaluate the immunogenicity of CD40L-dSIV in vitro. Expression levels of CD80, CD86, HLA-DR, and CD54 on DCs transduced with the dSIV incorporating CD40L (CD40L-dSIV) were significantly higher than on those transduced with dSIV. Moreover, CD40L-dSIV-transduced DCs expressed up to 10-fold more IL-12 than dSIV-transduced DCs. CD40L-dSIV-transduced DCs enhanced proliferation and gamma interferon secretion by naive T cells in an MLR. In addition, CD40L-dSIV-immunized mice exhibited stronger humoral and cell-mediated immune responses than dSIV-vaccinated animals. The results show that incorporating CD40L into the dSIV envelope significantly enhances immunogenicity. As a result, CD40L-dSIVs can be strong candidates for development of a safe and highly immunogenic AIDS vaccine.
Collapse
|
12
|
Yang DF, Qiu WH, Zhu HF, Lei P, Wen X, Dai H, Zhou W, Shen GX. CTLA4-Ig-modified dendritic cells inhibit lymphocyte-mediated alloimmune responses and prolong the islet graft survival in mice. Transpl Immunol 2008; 19:197-201. [PMID: 18667318 DOI: 10.1016/j.trim.2008.05.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2008] [Revised: 04/30/2008] [Accepted: 05/02/2008] [Indexed: 11/15/2022]
Abstract
The induction of antigen specific tolerance is critical for prevention and treatment of allograft rejection. In this study, we transfected CTLA4-Ig gene into dendritic cells (DCs), and investigated their effect on inhibition of lymphocyte activity in vitro and induction of immune tolerance on pancreatic islet allograft in mice. An IDDM C57BL/6 murine model induced by streptozotocin is as model mouse. The model mice were transplanted of the islet cells isolated from the BALB/c mice to their kidney capsules, and injected of CTLA4-Ig modified DCs (mDCs). The results showed that mDCs could significantly inhibit T lymphocyte proliferation and induce its apoptosis; whereas, unmodified DCs (umDCs) promoted the murine lymphocyte proliferation. Compared with injection of umDCs and IgG1 modified DCs, the injection of mDCs prolonged IDDM mice's allograft survival, and normalized their plasma glucose (PG) levels within 3 days and maintained over 2 weeks. The level of IFN-gamma was lower and the level of IL-4 was higher in mDCs treated recipient mice than that in control mice, it indicated that mDCs led to Th1/Th2 deviation. After 7 days of islet transplantation, HE stain of the renal specimens showed that the islets and kidneys were intact in structure, and islet cells numbers are increased in mDCs treated mice. Our studies suggest that DCs expressing CTLA4-Ig fusion protein can induce the immune tolerance to islet graft and prolong the allograft survival through the inhibition of T cell proliferation in allogeneic mice.
Collapse
Affiliation(s)
- Dao-Feng Yang
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, China
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Kapessidou P, Poulin L, Dumoutier L, Goldman M, Renauld JC, Braun MY. Interleukin-22 deficiency accelerates the rejection of full major histocompatibility complex-disparate heart allografts. Transplant Proc 2008; 40:1593-7. [PMID: 18589157 DOI: 10.1016/j.transproceed.2008.03.151] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2007] [Revised: 02/14/2008] [Accepted: 03/11/2008] [Indexed: 10/21/2022]
Abstract
Interleukin-22 (IL-22) was recently described as an effector cytokine produced by TH17 CD4(+) T lymphocytes that, cooperatively with IL-17, mediates IL-23-driven inflammation. Because there was experimental evidence for the role of IL-17 in acute rejection of vascularized allografts, we undertook the present study to assess the function of IL-22 in the process. There was an early transient expression of IL-22 in C57BL/6 mouse cardiac allografts (2-4 days posttransplantation) transplanted to BALB/c recipients. The main source of IL-22 among infiltrating leukocytes was cells expressing the macrophage/monocyte markers Mac3 and CD11b. T cells and granulocytes present in the rejected graft did not express IL-22. Surprisingly, the absence of IL-22 accelerated the rejection of fully histoincompatible hearts. Histology of rejected organs revealed the presence of intensive intragraft thrombosis and disseminated hemorrhagic necrosis. Taken together, these results demonstrated that IL-22 was not an effector lymphokine in cardiac allograft rejection, but early intragraft expression of the cytokine protected it from rejection.
Collapse
Affiliation(s)
- P Kapessidou
- Institute for Medical Immunology, Université Libre de Bruxelles (ULB), Gosselies, Belgium
| | | | | | | | | | | |
Collapse
|
14
|
IL-17 Production Elicited by Allo-Major Histocompatibility Complex Class II Recognition Depends on CD25posCD4pos T Cells. Transplantation 2008; 85:943-9. [DOI: 10.1097/tp.0b013e31816a5ae7] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
15
|
Li M, Zhang X, Zheng X, Lian D, Zhang ZX, Sun H, Suzuki M, Vladau C, Huang X, Xia X, Zhong R, Garcia B, Min WP. Tolerogenic dendritic cells transferring hyporesponsiveness and synergizing T regulatory cells in transplant tolerance. Int Immunol 2008; 20:285-293. [PMID: 18182381 DOI: 10.1093/intimm/dxm142] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2025] Open
Abstract
Dendritic cells are among the most potent antigen-presenting cells and are important in the development of both immunity and tolerance. Tolerogenic dendritic cell (Tol-DC) is a key factor in the induction and maintenance of tolerance during transplantation. However, the precise mechanism and direct evidence of in vivo immune modulation remain unclear. In the present study, we identified critical roles of immune modulation on transplant tolerance through interactions between Tol-DCs and regulatory T (Treg) cells. Tol-DCs remained in an immature state and were insensitive to maturation stimuli. Tol-DCs in tolerant recipients heightened the expression of indoleamine 2,3-dioxygenase (IDO) that induced allogeneic T-cell apoptosis. Adoptive transfer of Tol-DCs isolated from primary tolerant recipients resulted in augmentation of CD4(+)CD25(+)CTLA4(+) Treg cells and prolonged graft survival in secondary allogeneic heart transplantation and synergized with Treg cells to induce tolerance in secondary recipients. This study indicates that Tol-DC offers two functions during the process of tolerogenesis: suppression of anti-donor T-cell responses through production of IDO and interaction with Treg cells, which provides a framework for future research into tolerance induction.
Collapse
Affiliation(s)
- Mu Li
- Department of Surgery, University of Western Ontario, London, Ontario, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Li M, Zhang X, Zheng X, Lian D, Zhang ZX, Ge W, Yang J, Vladau C, Suzuki M, Chen D, Zhong R, Garcia B, Jevnikar AM, Min WP. Immune modulation and tolerance induction by RelB-silenced dendritic cells through RNA interference. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2007; 178:5480-5487. [PMID: 17442929 DOI: 10.4049/jimmunol.178.9.5480] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Dendritic cells (DC), the most potent APCs, can initiate the immune response or help induce immune tolerance, depending upon their level of maturation. DC maturation is associated with activation of the NF-kappaB pathway, and the primary NF-kappaB protein involved in DC maturation is RelB, which coordinates RelA/p50-mediated DC differentiation. In this study, we show that silencing RelB using small interfering RNA results in arrest of DC maturation with reduced expression of the MHC class II, CD80, and CD86. Functionally, RelB-silenced DC inhibited MLR, and inhibitory effects on alloreactive immune responses were in an Ag-specific fashion. RelB-silenced DC also displayed strong in vivo immune regulation. An inhibited Ag-specific response was seen after immunization with keyhole limpet hemocyanin-pulsed and RelB-silenced DC, due to the expansion of T regulatory cells. Administration of donor-derived RelB-silenced DC significantly prevented allograft rejection in murine heart transplantation. This study demonstrates for the first time that transplant tolerance can be induced by means of RNA interference using in vitro-generated tolerogenic DC.
Collapse
Affiliation(s)
- Mu Li
- Department of Surgery, University of Western Ontario, London Health Sciences Centre-University Campus, 339 Windermere Road, London, Ontario, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Miranda S, Litwin S, Barrientos G, Szereday L, Chuluyan E, Bartho JS, Arck PC, Blois SM. Dendritic Cells Therapy Confers a Protective Microenvironment in Murine Pregnancy. Scand J Immunol 2006; 64:493-9. [PMID: 17032241 DOI: 10.1111/j.1365-3083.2006.01841.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The fetal-placental unit is a semi-allograft and immunological recognition of pregnancy, together with the subsequent response of the maternal immune system, is necessary for a successful pregnancy. Dendritic cells (DC) show a biological plasticity that confers them special characteristics regulating both immunity and tolerance. Therapy employing DC proved to diminish the abortion in the DBA/2J-mated CBA/J females; however, the underlying mechanisms remain unknown. Here, we evaluated whether DC therapy influences the presence of immunoregulatory populations of cells at the fetal-maternal interface. To address this hypothesis, we analysed the pregnancy-protective CD8, gammadelta cell populations as well as transforming growth factor (TGF)-beta1 and progesterone-induced blocking factor (PIBF) expression at the fetal-maternal interface from abortion-prone female mice that had previously received adoptive transfer of syngeneic DC. Syngeneic DC therapy induced an increase in the number of CD8 and gammadelta cells. Additionally, an upregulation of TGF-beta1 and PIBF expression could be detected after DC transfer. We suggest that DC therapy differentially upregulates a regulatory/protective population of cells at the fetal-maternal interface. It is reasonable to assure that this mechanism would be responsible for the lower abortion rate.
Collapse
Affiliation(s)
- S Miranda
- Department of Microbiology, Biotechnology and Immunology, IDEHU (CONICET-University of Buenos Aires), Buenos Aires, Argentina
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Kapessidou Y, Habran C, Buonocore S, Flamand V, Barvais L, Goldman M, Braun MY. The replacement of graft endothelium by recipient-type cells conditions allograft rejection mediated by indirect pathway CD4(+) T cells. Transplantation 2006; 81:726-35. [PMID: 16534475 DOI: 10.1097/01.tp.0000184444.93108.d1] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Whereas the participation of alloreactive T cells sensitized by indirect allorecognition in graft rejection is well documented, the nature of recipient antigen presenting cells recognized by indirect pathway CD4(+) T cells within the graft has yet to be identified. The purpose of this study was to determine the role played by graft endothelium replacement in the immune recognition of cardiac allografts rejected by indirect pathway CD4(+) T cells. METHODS Transgenic RAG2(-/-) mice expressing I-A(b)-restricted male antigen H-Y-specific TcR were studied for their capacity to reject H-2(k) male cardiac allografts. Chronic vascular rejection in this model was due to the indirect recognition of H-Y antigen shed from H-2(k) male allograft and presented by the recipient's own I-A(b) APC to transgenic T cells. RESULTS Immunohistochemical analysis of rejected grafts revealed the presence of numerous microvascular endothelial cells (EC) that expressed recipient's I-A MHC class II molecules. This observation suggested that graft endothelium replacement by I-A(b)-positive cells of recipient origin could stimulate the rejection of male H-2(k) graft by I-A(b)--restricted H-Y--specific T cells. To investigate further this possibility, hearts from H-2(b)--into--H-2(k) irradiation bone marrow (BM) chimera were transplanted in transgenic recipients. A direct correlation was observed between the presence of I-A(b)-positive EC within myocardial microvessels and the induction of acute rejection of chimeric H-2(k) male cardiac allografts transplanted in transgenic recipients. CONCLUSIONS We conclude that graft endothelium replacement by recipient-type cells is required for the rejection of cardiac allograft mediated by indirect pathway alloreactive CD4(+) T cells.
Collapse
Affiliation(s)
- Yota Kapessidou
- Institute for Medical Immunology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | | | | | | | | | | | | |
Collapse
|
19
|
Pearl-Yafe M, Yolcu ES, Yaniv I, Stein J, Shirwan H, Askenasy N. The dual role of Fas-ligand as an injury effector and defense strategy in diabetes and islet transplantation. Bioessays 2006; 28:211-22. [PMID: 16435302 DOI: 10.1002/bies.20356] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The exact process that leads to the eruption of autoimmune reactions against beta cells and the evolution of diabetes is not fully understood. Macrophages and T cells may launch an initial immune reaction against the pancreatic islets of Langerhans, provoking inflammation and destructive insulitis. The information on the molecular mechanisms of the emergence of beta cell injury is controversial and points to possibly important roles for the perforin-granzyme, Fas-Fas-ligand (FasL) and tumor-necrosis-factor-mediated apoptotic pathways. FasL has several unique features that make it a potentially ideal immunomodulatory tool. Most important, FasL is selectively toxic to cytotoxic T cells and less harmful to regulatory T cells. This review discusses the intrinsic sensitivity of beta cells to FasL-mediated apoptosis, the conditions that underlie this beta cell sensitivity, and the feasibility of using FasL to arrest autoimmunity and prevent islet allograft rejection. In both the autoimmune and transplant settings, it is imperative to progress from the administration of nonspecific immunosuppressive therapy to the concept of beta-cell-specific immunomodulation. FasL evolves as a prime candidate for antigen-specific immunomodulation.
Collapse
Affiliation(s)
- Michal Pearl-Yafe
- Frankel Laboratory of Experimental Bone Marrow Transplantation, Department of Pediatric Hematology Oncology, Schneider Children's Medical Center of Israel, Israel
| | | | | | | | | | | |
Collapse
|
20
|
Laffont S, Coudert JD, Garidou L, Delpy L, Wiedemann A, Demur C, Coureau C, Guéry JC. CD8+ T-cell-mediated killing of donor dendritic cells prevents alloreactive T helper type-2 responses in vivo. Blood 2006; 108:2257-64. [PMID: 16449531 DOI: 10.1182/blood-2005-10-4059] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Accumulating evidence indicates that, in absence of CD8+ T-cell activation, CD4+ T-cell-mediated allograft rejection is associated with a dominant Th2-cell response and eosinophil infiltrates. In this study, we analyzed the mechanisms by which CD8+ T cells regulate alloreactive CD4+ T-cell priming and differentiation into interleukin 4 (IL-4)-producing cells. We showed that interferon gamma (IFN-gamma) production by CD8+ T cells was dispensable for the inhibition of Th2-cell development, as well as tissue eosinophilia and type 2 cytokine production in the rejected grafts. Since we noticed that CD8+ T cells not only suppressed Th2 differentiation, but also down-modulated the overall priming of alloreactive CD4+ T cells, we evaluated whether CD8+ T cells act by limiting the accumulation of donor-derived dendritic cells (DCs) in lymph nodes. We found that indeed, alloreactive CD8+ T cells rapidly eliminated allogeneic DCs from T-cell areas of draining lymph nodes, through a perforin-dependent mechanism. Thus, our data demonstrate that cytotoxic T lymphocyte (CTL)-mediated clearance of allogeneic DCs is a negative feedback mechanism that limits the duration of alloantigen presentation in draining lymph nodes, thereby modulating the amplitude and polarization of the primary alloreactive CD4+ T-cell responses.
Collapse
Affiliation(s)
- Sophie Laffont
- INSERM U563, CHU Purpan, Place du Dr Baylac, 31 300 Toulouse, France
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Surquin M, Le Moine A, Flamand V, Rombaut K, Demoor FX, Salmon I, Goldman M, Abramowicz D. IL-4 deficiency prevents eosinophilic rejection and uncovers a role for neutrophils in the rejection of MHC class II disparate skin grafts. Transplantation 2006; 80:1485-92. [PMID: 16340795 DOI: 10.1097/01.tp.0000176486.01697.3f] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Acute rejection of MHC class II-disparate bm12 skin grafts by C57BL/6 recipient mice is characterized by massive graft infiltration by eosinophils, together with increased intragraft amounts of IL-4 and IL-5 mRNA. IL-5 blockade prevents the intragraft eosinophil infiltration and prolongs the survival of skin allografts. As the differentiation of T cell precursors into Th2 cells is largely driven by IL-4, we investigated the role of IL-4 in MHC class II-disparate allograft rejection. METHODS We performed skin grafts from MHC class II incompatible bm12 mice into wild-type C57BL/6 mice (IL-4) or C57BL/6 IL-4 deficient mice (IL-4). Graft survival, in vitro T cell reactivity, and histology were compared. RESULTS We observed that 50% of IL-4 mice rapidly rejected their bm12 allograft, whereas the other 50% retained their graft 60 days after transplantation. Histological examination of bm12 allografts retained by IL-4 mice showed a normal appearance with no inflammatory infiltrate and no eosinophils. Among IL-4 mice that acutely rejected their bm12 skin graft, we observed a dense polymorphonuclear infiltrate. The depletion of neutrophils significantly prolonged bm12 graft survival. CONCLUSIONS Eosinophil infiltrates, typical of MHC class II disparate acute skin graft rejection, are critically dependent on the availability of IL-4. IL-4 mice reject MHC class II disparate skin grafts by a pathway of rejection where neutrophils play a direct causal role.
Collapse
|
22
|
Moore F, Buonocore S, Paulart F, Thielemans K, Goldman M, Flamand V. Unexpected effects of viral interleukin-10-secreting dendritic cells in vivo: preferential inhibition of TH2 responses. Transplant Proc 2005; 36:3260-6. [PMID: 15686742 DOI: 10.1016/j.transproceed.2004.10.039] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Viral interleukin (IL)-10 (vIL-10) has been widely described as an immunoregulatory cytokine that does not possess the T-cell costimulatory activities of cellular IL-10; it was therefore believed to be a more potent tolerogenic mediator. The immunosuppressive properties of this cytokine are partly attributed to its capacity to render dendritic cells (DCs) unable to undergo full maturation and to activate T cells. We reported here that myeloid DCs retrovirally transduced with vIL-10 had an impaired production of IL-12 and a decreased expression of MHC class II molecules but had minor defects in costimulatory molecule expression and no alteration on CCR5 and CCR7 expression. In mixed leukocyte reaction, vIL-10-transduced C57BL/6 bm12 (MHC class II mismatch) DCs had a reduced capacity to stimulate C57BL/6 wild-type CD4+ T-cell proliferation. We show that bm12 vIL-10-transduced DC administration in CD8-/- C57BL/6 mice promoted IFN-gamma production, down-regulated TH2-type cytokine production, and did not induce skin graft tolerance. These findings suggest that vIL-10-transduced DC may surprisingly facilitate Th1-type inflammatory responses in vivo.
Collapse
Affiliation(s)
- F Moore
- Institute for Medical Immunology, Université Libre de Bruxelles
| | | | | | | | | | | |
Collapse
|
23
|
Benghiat FS, Graca L, Braun MY, Detienne S, Moore F, Buonocore S, Flamand V, Waldmann H, Goldman M, Le Moine A. Critical influence of natural regulatory CD25+ T cells on the fate of allografts in the absence of immunosuppression. Transplantation 2005; 79:648-54. [PMID: 15785370 DOI: 10.1097/01.tp.0000155179.61445.78] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
BACKGROUND Allografts are occasionally accepted in the absence of immunosuppression. Because naturally occurring CD4(+)CD25(+) regulatory T cells (natural CD25(+) Treg cells) have been shown to inhibit allograft rejection, we investigated their influence on the outcome of allografts in nonimmunosuppressed mouse recipients. METHODS We compared survival times of male CBA/Ca skin grafts in female CBA/Ca recipients expressing a transgenic anti-HY T-cell receptor on a RAG-1(+/+) (A1[M]RAG+) or a RAG-1(-/-) (A1[M]RAG-) background. Depletion of natural CD25(+) Treg cells in A1[M]RAG+ mice was achieved by in vivo administration of the PC61 monoclonal antibody. The influence of natural CD25(+) Treg cells on the fate of major histocompatibility complex class II-mismatched (C57BL/6X bm12)F1 skin or bm12 heart transplants in C57BL/6 recipients was also assessed. Finally, we investigated the impact of natural CD25(+) Treg cells on the production of T-helper (Th)1 and Th2 cytokines in mixed lymphocyte cultures between C57BL/6 CD4(+) CD25(-) T cells as responders and bm12 or (C57BL/6X bm12)F1 antigen-presenting cells as stimulators. RESULTS Male allografts were spontaneously accepted by female A1(M)RAG+ mice but readily rejected by female A1(M)RAG+ mice depleted of natural CD25(+) Treg cells by pretreatment with the PC61 monoclonal antibody. Depletion of CD25(+) Treg cells also enhanced eosinophil-determined rejection of (C57BL/6X bm12)F1 skin grafts or bm12 cardiac grafts in C57BL/6 recipients. Finally, natural CD25(+) Treg cells inhibited the production of interleukin (IL)-2, interferon-gamma, IL-5, and IL-13 in mixed lymphocyte culture in a dose-dependent manner. CONCLUSION Natural CD25(+) Treg cells control Th1- and Th2-type allohelper T-cell responses and thereby influence the fate of allografts in nonimmunosuppressed recipients.
Collapse
MESH Headings
- Animals
- Cytokines/immunology
- Cytokines/metabolism
- Female
- Graft Rejection/immunology
- Graft Survival/immunology
- Heart Transplantation/immunology
- Heart Transplantation/pathology
- Immunosuppression Therapy
- Lymphocyte Culture Test, Mixed
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Rats
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Interleukin-2/immunology
- Receptors, Interleukin-2/metabolism
- Skin Transplantation/immunology
- Skin Transplantation/pathology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Th1 Cells/immunology
- Th1 Cells/metabolism
- Th2 Cells/immunology
- Th2 Cells/metabolism
- Transplantation
- Transplantation, Homologous/immunology
Collapse
Affiliation(s)
- Fleur Samantha Benghiat
- Institute for Medical Immunology, Université Libre de Bruxelles, 8, rue Adrienne Bolland, B-6041 Charleroi (Gosselies), Belgium
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Taner T, Hackstein H, Wang Z, Morelli AE, Thomson AW. Rapamycin-treated, alloantigen-pulsed host dendritic cells induce ag-specific T cell regulation and prolong graft survival. Am J Transplant 2005; 5:228-36. [PMID: 15643982 DOI: 10.1046/j.1600-6143.2004.00673.x] [Citation(s) in RCA: 207] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Tolerogenic properties of dendritic cells (DC), particularly those in the immature state, and their therapeutic potential are increasingly being recognized. Among several distinct approaches to generate stably immature DC, pharmacologic manipulation stands out as a promising and clinically applicable option. We have shown recently that the immunophilin ligand rapamycin (Rapa) can inhibit DC maturation and their effector functions. Here, we examined the impact of Rapa exposure on subsequent alloantigen (Ag) presentation by myeloid DC via the indirect pathway. Rapa-treated, allogeneic lysate-pulsed host DC (Rapa-DC) were inferior stimulators of syngeneic T cells, compared to lysate-pulsed control DC. Rapa exposure did not block alloAg uptake by DC nor impair their in vivo homing to splenic T cell areas after adoptive transfer. T cells primed by Rapa-treated, alloAg-pulsed DC showed decreased capacity to produce IL-2 and IFNgamma, and were hyporesponsive to subsequent challenge via both the direct and indirect pathways, in an Ag-specific manner. When infused 1 week before transplantation, these Rapa-DC significantly prolonged alloAg-specific heart graft survival. This effect was reversed by systemic IL-2 administration but enhanced by either repeated infusion of the cells or a short post-transplant course of FK506. These therapeutic effects, achieved by targeting both major pathways of allorecognition, provide the basis for a clinically applicable strategy to suppress graft rejection.
Collapse
Affiliation(s)
- Timuçin Taner
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh, PA, USA
| | | | | | | | | |
Collapse
|
25
|
Abstract
There has been tremendous progress in our understanding of the mechanisms mediating allograft tolerance, which have been revealed to be far more complex and regulated than hitherto suspected. New results have enriched our understanding of the relative contributions of the direct and indirect pathways to immunity and tolerance over time. The role of central tolerance has been expanded with the surprising discovery of "ectopic" or "promiscuous" antigens expressed by medullary thymic epithelial cells, and the function of the thymus in generating naturally occurring CD4+ CD25+ regulatory T cells. In the periphery, it is increasingly appreciated that tolerance is a highly active process, with tolerogenic dendritic cells and regulatory T cells being the major players. However, the challenge of understanding the complex interactions regulating the dynamic balance between immunity and tolerance are formidable, and new tools from the more formal disciplines of nonlinear dynamics and systems engineering may help provide insight. Although many hurdles remain, the progress in elucidating the basic mechanisms of tolerance is rapidly being translated into clinical trials and provides grounds for optimism that clinical tolerance will eventually become a reality.
Collapse
Affiliation(s)
- C Chan
- Department of Immunology, Division of Medicine, Imperial College London, London, UK
| | | | | |
Collapse
|
26
|
Ichim TE, Zhong R, Min WP. Prevention of allograft rejection by in vitro generated tolerogenic dendritic cells. Transpl Immunol 2003; 11:295-306. [PMID: 12967783 DOI: 10.1016/s0966-3274(03)00048-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Achieving immunological tolerance in transplantation has been a long sought-after goal since the 1960s. It is, therefore, interesting that the dendritic cells (DC), which are classically known as the most potent stimulators of T cell activation, are now also considered putative tools for tolerance induction. In line with this, much work has been performed using DC for vaccination and immune stimulation. Recently, great interest has been generated regarding the ability of DC to act as immune regulatory cells. Specific subsets of DC or immature DC (iDC) appear to be responsible for maintaining self-tolerance. In this review we will highlight our efforts at elucidating the contribution of DC in transplant tolerant in mice. Specifically, four strategies will be outlined that are currently being used for the generation of DC that have tolerogenic properties in the prevention of allograft rejection. The present study demonstrates that modulated iDC with blunted T cell stimulatory or antigen presentation abilities can afford transplant tolerance by minimizing T cell activation and proinflammatory cytokine production. Moreover, in an alternate strategy, normally matured DC have also been modulated such that alloreactive T cells are specifically targeted for deletion.
Collapse
Affiliation(s)
- Thomas E Ichim
- Department of Surgery, University of Western Ontario, London, Ont., Canada
| | | | | |
Collapse
|
27
|
Fresnay S, Garnache-Ottou F, Plumas J, Seilles E, Tiberghien P, Saas P. Can tolerogenic dendritic cells help to modulate allo-immune responses in the setting of hematopoietic cell transplantation? Transpl Immunol 2003; 11:259-66. [PMID: 12967779 DOI: 10.1016/s0966-3274(03)00053-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Recent evidence suggests that recipient as well as donor dendritic cell (DC) subsets are implicated in hematopoietic engraftment, graft-vs.-host disease occurrence, immune reconstitution and graft-vs.-leukemia effects observed after allogeneic hematopoietic cell transplantation. Although further data are needed to better understand the precise role of different DC subsets, strategies based on their manipulation to obtain tolerogenic DC can be envisaged. Here, we propose that DC blocked in an immature stage, using immunosuppressive agents, or lymphoid DC can be adequate candidates to control the alloreactive conflict post-allograft.
Collapse
Affiliation(s)
- Stéphanie Fresnay
- INSERM E0119, Etablissement Français du Sang Bourgogne Franche-Comté, MEN EA2284, Université de Franche-Comté, Besancon, France
| | | | | | | | | | | |
Collapse
|
28
|
Toungouz M, Donckier V, Goldman M. Tolerance induction in clinical transplantation: the pending questions. Transplantation 2003; 75:58S-60S. [PMID: 12819494 DOI: 10.1097/01.tp.0000067955.60639.d5] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Despite the dramatic improvement of early graft survival due to the introduction of cyclosporin, late graft loss caused by chronic rejection and the lethal consequences of long-term immunosuppression, such as infection and cancer, remain major concerns for the transplantation community. Tolerance induction would avoid these complications. The ways to go are controversial, reflecting the redundancy of rejection pathways. They include the induction of central tolerance by establishment of mixed chimerism through hematopoietic stem cell transplantation and the induction of "operational tolerance" through immunodeviation involving dendritic or regulatory T cells. Major advances have been made in animal models exploring these strategies and, some preliminary data are even now available in humans, allowing the initiation of pilot clinical trials. In this article, we discuss the key questions that these trials will have to address.
Collapse
Affiliation(s)
- Michel Toungouz
- Department of Immunology-Hematology-Transfusion, Hôpital Erasme, Universite Libre de Bruxelles, Belgium.
| | | | | |
Collapse
|