1
|
Andrade MS, Young JS, Pollard JM, Yin D, Alegre ML, Chong AS. Linked sensitization by memory CD4+ T cells prevents costimulation blockade–induced transplantation tolerance. JCI Insight 2022; 7:159205. [PMID: 35674134 PMCID: PMC9220839 DOI: 10.1172/jci.insight.159205] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 04/22/2022] [Indexed: 11/17/2022] Open
Abstract
Dominant infectious tolerance explains how brief tolerance-inducing therapies result in lifelong tolerance to donor antigens and “linked” third-party antigens, while recipient sensitization and ensuing immunological memory prevent the successful induction of transplant tolerance. In this study, we juxtapose these 2 concepts to test whether mechanisms of dominant infectious tolerance can control a limited repertoire of memory T and B cells. We show that sensitization to a single donor antigen is sufficient to prevent stable transplant tolerance, rendering it unstable. Mechanistic studies revealed that recall antibody responses and memory CD8+ T cell expansion were initially controlled, but memory CD4+Foxp3– T cell (Tconv) responses were not. Remarkably, naive donor-specific Tconvs at tolerance induction also acquired a resistance to tolerance, proliferating and acquiring a phenotype similar to memory Tconvs. This phenomenon of “linked sensitization” underscores the challenges of reprogramming a primed immune response toward tolerance and identifies a potential therapeutic checkpoint for synergizing with costimulation blockade to achieve transplant tolerance in the clinic.
Collapse
|
2
|
Waldmann H. Regulatory T cells and transplantation tolerance: Emerging from the darkness? Eur J Immunol 2021; 51:1580-1591. [PMID: 33961297 DOI: 10.1002/eji.202048795] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 03/29/2021] [Accepted: 05/05/2021] [Indexed: 12/11/2022]
Abstract
The field of tissue transplantation has revolutionized the treatment of patients with failing organs. Its success, thus far, has depended on combinations of immunosuppressive drugs that damp host immunity, while also imposing numerous unwanted side-effects. There is a longstanding recognition that better treatment outcomes, will come from replacing these drugs, fully or in part, by taking advantage of tractable physiological mechanisms of self-tolerance. The past 50 years have seen many advances in the field of self-tolerance, but perhaps, the most tractable of these has been the more recent discovery of a subset T-cells (Treg) whose role is to regulate or damp immunity. This article is intended to first provide the reader with some historical background to explain why we have been slow to identify these cells, despite numerous clues to their existence, and also to indicate how little we know about how they achieve their regulatory function in averting transplant rejection. However, as is often the case in immunology, the therapeutic needs often dictate that our advances move to translation even before detailed explanations of the science are available. The final part of the article will briefly summarize how Treg are being harnessed as agents to interface with or perhaps, replace current drug combinations.
Collapse
Affiliation(s)
- Herman Waldmann
- Sir William Dunn School, University of Oxford, Oxford, OX13RE, UK
| |
Collapse
|
3
|
Rojas C, Campos-Mora M, Cárcamo I, Villalón N, Elhusseiny A, Contreras-Kallens P, Refisch A, Gálvez-Jirón F, Emparán I, Montoya-Riveros A, Vernal R, Pino-Lagos K. T regulatory cells-derived extracellular vesicles and their contribution to the generation of immune tolerance. J Leukoc Biol 2020; 108:813-824. [PMID: 32531824 DOI: 10.1002/jlb.3mr0420-533rr] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 04/15/2020] [Accepted: 04/27/2020] [Indexed: 12/11/2022] Open
Abstract
T regulatory (Treg) cells have a major role in the maintenance of immune tolerance against self and foreign antigens through the control of harmful inflammation. Treg cells exert immunosuppressive function by several mechanisms, which can be distinguished as contact dependent or independent. Recently, the secretion of extracellular vesicles (EVs) by Treg cells has been reported as a novel suppressive mechanism capable of modulating immunity in a cell-contact independent and targeted manner, which has been identified in different pathologic scenarios. EVs are cell-derived membranous structures involved in physiologic and pathologic processes through protein, lipid, and genetic material exchange, which allow intercellular communication. In this review, we revise and discuss current knowledge on Treg cells-mediated immune tolerance giving special attention to the production and release of EVs. Multiple studies support that Treg cells-derived EVs represent a refined intercellular exchange device with the capacity of modulating immune responses, thus creating a tolerogenic microenvironment in a cell-free manner. The mechanisms proposed encompass miRNAs-induced gene silencing, the action of surface proteins and the transmission of enzymes. These observations gain relevance by the fact that Treg cells are susceptible to converting into effector T cells after exposition to inflammatory environments. Yet, in contrast to their cells of origin, EVs are unlikely to be modified under inflammatory conditions, highlighting the advantage of their use. Moreover, we speculate in the possibility that Treg cells may contribute to infectious tolerance via vesicle secretion, intervening with CD4+ T cells differentiation and/or stability.
Collapse
Affiliation(s)
- Carolina Rojas
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Las Condes, Santiago, Chile.,Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Las Condes, Santiago, Chile
| | - Mauricio Campos-Mora
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Las Condes, Santiago, Chile
| | - Ignacio Cárcamo
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Las Condes, Santiago, Chile
| | - Natalia Villalón
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Las Condes, Santiago, Chile
| | - Ahmed Elhusseiny
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Las Condes, Santiago, Chile
| | - Pamina Contreras-Kallens
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Las Condes, Santiago, Chile
| | - Aarón Refisch
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Las Condes, Santiago, Chile
| | - Felipe Gálvez-Jirón
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Las Condes, Santiago, Chile
| | - Ivana Emparán
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Las Condes, Santiago, Chile
| | - Andro Montoya-Riveros
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Las Condes, Santiago, Chile
| | - Rolando Vernal
- Periodontal Biology Laboratory, Faculty of Dentistry, Universidad de Chile, Las Condes, Santiago, Chile
| | - Karina Pino-Lagos
- Centro de Investigación Biomédica, Facultad de Medicina, Universidad de los Andes, Las Condes, Santiago, Chile
| |
Collapse
|
4
|
Waldmann H, Graca L. Infectious tolerance. What are we missing? Cell Immunol 2020; 354:104152. [PMID: 32585469 DOI: 10.1016/j.cellimm.2020.104152] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 06/10/2020] [Accepted: 06/10/2020] [Indexed: 11/16/2022]
Affiliation(s)
- Herman Waldmann
- Sir William Dunn School of Pathology, South Parks Road, Oxford OX13RE, UK.
| | - Luis Graca
- Instituto de Medecina Molecular, Faculdade de medicina da Universidade de Lisboa, Avenida professor Egas Moniz, 1649-028 Lisboa, Portugal.
| |
Collapse
|
5
|
Xu N, Ruan G, Liu W, Hu C, Huang A, Zeng Z, Luo S, Zhang Z, Fan M, Ye F, Xi T, Xing Y. Vaccine-induced gastric CD4 + tissue-resident memory T cells proliferate in situ to amplify immune response against Helicobacter pylori insult. Helicobacter 2019; 24:e12652. [PMID: 31414552 DOI: 10.1111/hel.12652] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 07/03/2019] [Accepted: 07/09/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Tissue-resident memory T cells accelerate the clearance of pathogens during recall response. However, whether CD4+ TRM cells themselves can provide gastric immunity is unclear. MATERIALS AND METHODS We established a parabiosis model between the enhanced green fluorescent protein and wild-type mice that the circulation system was shared, and the wild-type partner was vaccinated with H pylori vaccine composed of CCF and silk fibroin in gastric subserous layer to induce gastric EGFP+ CD4+ TRM cells. Antigen-specific EGFP+ CD4+ T cells and proliferous TRM cells were analyzed by flow cytometry. The colonization of H pylori was detected by quantitative real-time PCR. EGFP+ CD4+ TRM cells and the inflammation of the stomach were observed by histology. RESULTS A parabiosis animal model was employed to identify the cells that introduced by vaccination in GSL. Antigen-specific EGFP+ CD4+ T cells could be detected at day 7 post-vaccination. Thirty days later, EGFP+ CD4+ TRM cells were established with a phenotype of CD69+ CD103- . Of note, we found that when circulating lymphocytes were depleted by FTY720 administration, these TRM cells could proliferate in situ and differentiate into effector Th1 cells after H pylori challenge. A decrease in H pylori colonization was observed in the vaccinated mice but not unvaccinated mice. Further, we found that although FTY720 was administrated, mounted pro-inflammatory myeloid cells still emerged in the stomach of the vaccinated mice, which might contribute to the reduction of H pylori colonization. CONCLUSIONS Our study reveals that H pylori vaccine-induced CD4+ TRM cells can proliferate and differentiate in situ to enhance gastric local immunity during recall response.
Collapse
Affiliation(s)
- Ningyin Xu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Guojing Ruan
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Wei Liu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Chupeng Hu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - An Huang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Zhiqin Zeng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Shuanghui Luo
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Zhenxing Zhang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Menghui Fan
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Feng Ye
- Department of Gastroenterology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Tao Xi
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Yingying Xing
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
6
|
Arefanian H, Tredget EB, Mok DCM, Ramji Q, Rafati S, Rodriguez-Barbosa J, Korbutt GS, Rajotte RV, Gill RG, Rayat GR. Porcine Islet-Specific Tolerance Induced by the Combination of Anti-LFA-1 and Anti-CD154 mAbs is Dependent on PD-1. Cell Transplant 2016; 25:327-42. [DOI: 10.3727/096368915x688506] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
We previously demonstrated that short-term administration of a combination of anti-LFA-1 and anti-CD154 monoclonal antibodies (mAbs) induces tolerance to neonatal porcine islet (NPI) xenografts that is mediated by regulatory T cells (Tregs) in B6 mice. In this study, we examined whether the coinhibitory molecule PD-1 is required for the induction and maintenance of tolerance to NPI xenografts. We also determined whether tolerance to NPI xenografts could be extended to allogeneic mouse or xenogeneic rat islet grafts since we previously demonstrated that tolerance to NPI xenografts could be extended to second-party NPI xenografts. Finally, we determined whether tolerance to NPI xenografts could be extended to allogeneic mouse or second-party porcine skin grafts. Diabetic B6 mice were transplanted with 2,000 NPIs under the kidney capsule and treated with short-term administration of a combination of anti-LFA-1 and anti-CD154 mAbs. Some of these mice were also treated simultaneously with anti-PD-1 mAb at >150 days posttransplantation. Spleen cells from some of the tolerant B6 mice were used for proliferation assays or were injected into B6 rag-/- mice with established islet grafts from allogeneic or xenogeneic donors. All B6 mice treated with anti-LFA-1 and anti-CD154 mAbs achieved and maintained normoglycemia until the end of the study; however, some mice that were treated with anti-PD-1 mAb became diabetic. All B6 rag-/- mouse recipients of first- and second-party NPIs maintained normoglycemia after reconstitution with spleen cells from tolerant B6 mice, while all B6 rag-/- mouse recipients of allogeneic mouse or xenogeneic rat islets rejected their grafts after cell reconstitution. Tolerant B6 mice rejected their allogeneic mouse or xenogeneic second-party porcine skin grafts while remaining normoglycemic until the end of the study. These results show that porcine islet-specific tolerance is dependent on PD-1, which could not be extended to skin grafts.
Collapse
Affiliation(s)
- Hossein Arefanian
- Alberta Diabetes Institute, Surgical-Medical Research Institute, Department of Surgery, Faculty of Medicine and Dentistry University of Alberta, Edmonton, Alberta, Canada
- Endocrinology and Metabolism Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Pancreatic Islet Biology and Transplantation Unit, Dasman Diabetes Institute, Kuwait, Dasman, Kuwait
| | - Eric B. Tredget
- Alberta Diabetes Institute, Surgical-Medical Research Institute, Department of Surgery, Faculty of Medicine and Dentistry University of Alberta, Edmonton, Alberta, Canada
| | - Dereck C. M. Mok
- Alberta Diabetes Institute, Surgical-Medical Research Institute, Department of Surgery, Faculty of Medicine and Dentistry University of Alberta, Edmonton, Alberta, Canada
| | - Qahir Ramji
- Alberta Diabetes Institute, Surgical-Medical Research Institute, Department of Surgery, Faculty of Medicine and Dentistry University of Alberta, Edmonton, Alberta, Canada
| | - Shahin Rafati
- Alberta Diabetes Institute, Surgical-Medical Research Institute, Department of Surgery, Faculty of Medicine and Dentistry University of Alberta, Edmonton, Alberta, Canada
| | - Jose Rodriguez-Barbosa
- Institute of Biomedicine (Immunobiology), University of Leon, Campus de Vegazana s/n, Leon, Spain
| | - Gregory S. Korbutt
- Alberta Diabetes Institute, Surgical-Medical Research Institute, Department of Surgery, Faculty of Medicine and Dentistry University of Alberta, Edmonton, Alberta, Canada
| | - Ray V. Rajotte
- Alberta Diabetes Institute, Surgical-Medical Research Institute, Department of Surgery, Faculty of Medicine and Dentistry University of Alberta, Edmonton, Alberta, Canada
| | - Ron G. Gill
- Barbara Davis Center for Childhood Diabetes, University of Colorado, Aurora, CO, USA
| | - Gina R. Rayat
- Alberta Diabetes Institute, Surgical-Medical Research Institute, Department of Surgery, Faculty of Medicine and Dentistry University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
7
|
Villani V, Yamada K, Scalea JR, Gillon BC, Arn JS, Sekijima M, Tasaki M, Cormack TA, Moran SG, Torabi R, Shimizu A, Sachs DH. Adoptive Transfer of Renal Allograft Tolerance in a Large Animal Model. Am J Transplant 2016; 16:317-24. [PMID: 26260215 PMCID: PMC4718904 DOI: 10.1111/ajt.13424] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 05/19/2015] [Accepted: 06/11/2015] [Indexed: 01/25/2023]
Abstract
Our recent studies in an inbred swine model demonstrated that both peripheral and intra-graft regulatory cells were required for the adoptive transfer of tolerance to a second, naïve donor-matched kidney. Here, we have asked whether both peripheral and intra-graft regulatory elements are required for adoptive transfer of tolerance when only a long-term tolerant (LTT) kidney is transplanted. Nine highly-inbred swine underwent a tolerance-inducing regimen to prepare LTT kidney grafts which were then transplanted to histocompatible recipients, with or without the peripheral cell populations required for adoptive transfer of tolerance to a naïve kidney. In contrast to our previous studies, tolerance of the LTT kidney transplants alone was achieved without transfer of additional peripheral cells and without strategies to increase the number/potency of regulatory T cells in the donor. This tolerance was systemic, since most subsequent, donor-matched challenge kidney grafts were accepted. These results confirm the presence of a potent tolerance-inducing and/or tolerance-maintaining cell population within LTT renal allografts. They suggest further that additional peripheral tolerance mechanisms, required for adoptive transfer of tolerance to a naïve donor-matched kidney, depend on peripheral cells that, if not transferred with the LTT kidney, require time to develop in the adoptive host.
Collapse
Affiliation(s)
- V Villani
- Transplantation Biology Research Center (TBRC) Laboratories, Center for Transplantation Sciences (CTS), Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - K Yamada
- Transplantation Biology Research Center (TBRC) Laboratories, Center for Transplantation Sciences (CTS), Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - J R Scalea
- Transplantation Biology Research Center (TBRC) Laboratories, Center for Transplantation Sciences (CTS), Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - B C Gillon
- Transplantation Biology Research Center (TBRC) Laboratories, Center for Transplantation Sciences (CTS), Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - J S Arn
- Transplantation Biology Research Center (TBRC) Laboratories, Center for Transplantation Sciences (CTS), Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - M Sekijima
- Transplantation Biology Research Center (TBRC) Laboratories, Center for Transplantation Sciences (CTS), Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - M Tasaki
- Transplantation Biology Research Center (TBRC) Laboratories, Center for Transplantation Sciences (CTS), Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - T A Cormack
- Transplantation Biology Research Center (TBRC) Laboratories, Center for Transplantation Sciences (CTS), Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - S G Moran
- Transplantation Biology Research Center (TBRC) Laboratories, Center for Transplantation Sciences (CTS), Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - R Torabi
- Transplantation Biology Research Center (TBRC) Laboratories, Center for Transplantation Sciences (CTS), Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - A Shimizu
- Transplantation Biology Research Center (TBRC) Laboratories, Center for Transplantation Sciences (CTS), Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - D H Sachs
- Transplantation Biology Research Center (TBRC) Laboratories, Center for Transplantation Sciences (CTS), Massachusetts General Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
8
|
Plain KM, Verma ND, Tran GT, Nomura M, Boyd R, Robinson CM, Hodgkinson SJ, Hall BM. Cytokines affecting CD4(+) T regulatory cells in transplant tolerance. Interleukin-4 does not maintain alloantigen specific CD4(+)CD25(+) Treg. Transpl Immunol 2013; 29:51-9. [PMID: 24139939 DOI: 10.1016/j.trim.2013.10.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Revised: 10/03/2013] [Accepted: 10/03/2013] [Indexed: 12/31/2022]
Abstract
IL-4 is thought to promote induction of transplantation tolerance and alloantigen-specific CD4(+)CD25(+) T regulatory cells (Treg). This study examined the effect of IL-4 on the induction and maintenance of the CD4(+) T regulatory cells (Treg) that mediate transplantation tolerance. Tolerance was induced in DA rats with PVG heterotopic cardiac allografts by a short course of cyclosporine. Naïve and tolerant lymphocytes, including the CD4(+) and CD4(+)CD25(+) T cell subsets, were assayed in mixed lymphocyte cultures with or without recombinant (r)IL-4 or other cytokines. The proliferation, cell surface and cytokine phenotype of these cells was examined, as was their capacity to adoptively transfer tolerance. rIL-4 enhanced the proliferation of naïve and tolerant lymphoid cells, including CD4(+) and CD4(+)CD25(+) T cells, but this was not alloantigen specific. Naïve or tolerant CD4(+) T cells cultured with rIL-4 and donor PVG antigen effected rapid graft rejection, even though before culture tolerant CD4(+) T cells transferred antigen-specific tolerance. These rIL-4 cultured CD4(+) T cells had a phenotype consistent with activated CD4(+)CD25(+)FoxP3(-) Th2 cells. While naïve natural CD4(+)CD25(+) T cells (nTreg) cultured with alloantigen and rIL-4 had enhanced proliferation and capacity to suppress rejection in vivo, the culture of tolerant CD4(+)CD25(+) T cells with alloantigen and rIL-4 could not sustain their proliferation against specific donor, nor their capacity to transfer tolerance to specific donor allograft. Thus, IL-4 promotes both regulatory and effector T cells early in the immune response, but once alloimmune tolerance is established, IL-4 promoted the activation of effector cells to mediate rejection and did not support alloantigen-specific Treg that could transfer specific tolerance.
Collapse
Affiliation(s)
- Karren M Plain
- Immune Tolerance Laboratory Faculty of Medicine, University of New South Wales, Sydney, Australia
| | | | | | | | | | | | | | | |
Collapse
|
9
|
Long-term prevention of chronic allograft rejection by regulatory T-cell immunotherapy involves host Foxp3-expressing T cells. Blood 2013; 121:4303-10. [DOI: 10.1182/blood-2012-08-452037] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Key Points
Administration of donor-specific regulatory T cells prevents chronic rejection of BM and skin allografts in the mouse. Injected regulatory T cells induce the emergence of host regulatory T cells with similar specificity thus ensuring persistence of tolerance.
Collapse
|
10
|
Yuan Q, Hong S, Shi B, Kers J, Li Z, Pei X, Xu L, Wei X, Cai M. CD4(+)CD25(-)Nrp1(+) T cells synergize with rapamycin to prevent murine cardiac allorejection in immunocompetent recipients. PLoS One 2013; 8:e61151. [PMID: 23577203 PMCID: PMC3618334 DOI: 10.1371/journal.pone.0061151] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Accepted: 03/06/2013] [Indexed: 12/18/2022] Open
Abstract
Besides CD4+CD25+Foxp3+ regulatory T cells (Tregs), other immunosuppressive T cells also participated in the regulation of immune tolerance. Reportedly, neuropilin-1 (Nrp1) might be one of the molecules by which regulatory cells exert their suppressive effects. Indeed, CD4+CD25−Nrp1+ T cells exhibit potent suppressive function in autoimmune inflammatory responses. Here we investigated the specific role of CD4+CD25−Nrp1+ T cells in the setting of the transplant immune response. Through MLR assays, we found that CD4+CD25−Nrp1+ T cells suppressed the proliferation of naive CD4+CD25− T cells activated by allogeneic antigen-stimulation. Adoptive transfer of CD4+CD25−Nrp1+ T cells synergized with rapamycin to induce long-term graft survival in fully MHC-mismatched murine heart transplantation, which was associated with decreased IFN-γ, IL-17 and increased IL-10, TGF-β, Foxp3 and Nrp1 expression in the grafts. Importantly, our data indicated that CD4+CD25−Nrp1+ T cell transfer augments the accumulation of Tregs in the recipient, and creates conditions that favored induction of hyporesponsiveness of the T effector cells. In conclusion, this translational study indicates the possible therapeutic potential of CD4+CD25−Nrp1+ T cells in preventing allorejection. CD4+Nrp1+ T cells might therefore be used in bulk as a population of immunosuppressive cells with more beneficial properties concerning ex vivo isolation as compared to Foxp3+ Tregs.
Collapse
Affiliation(s)
- Qing Yuan
- Organ Transplant Center, Organ Transplant Institution of PLA, The 309th Hospital of PLA, Beijing, China
| | - Shanjuan Hong
- National Key Laboratory of Medical Immunology, Second Military Medical University, Shanghai, China
| | - Bingyi Shi
- Organ Transplant Center, Organ Transplant Institution of PLA, The 309th Hospital of PLA, Beijing, China
| | - Jesper Kers
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Zhouli Li
- Organ Transplant Center, Organ Transplant Institution of PLA, The 309th Hospital of PLA, Beijing, China
| | - Xiangke Pei
- Organ Transplant Center, The 281th Hospital of PLA, Qinhuangdao, Hebei, China
| | - Liang Xu
- Organ Transplant Center, Organ Transplant Institution of PLA, The 309th Hospital of PLA, Beijing, China
| | - Xing Wei
- Organ Transplant Center, Organ Transplant Institution of PLA, The 309th Hospital of PLA, Beijing, China
| | - Ming Cai
- Organ Transplant Center, Organ Transplant Institution of PLA, The 309th Hospital of PLA, Beijing, China
- * E-mail:
| |
Collapse
|
11
|
Sagoo P, Lombardi G, Lechler RI. Relevance of regulatory T cell promotion of donor-specific tolerance in solid organ transplantation. Front Immunol 2012; 3:184. [PMID: 22811678 PMCID: PMC3395995 DOI: 10.3389/fimmu.2012.00184] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Accepted: 06/14/2012] [Indexed: 01/29/2023] Open
Abstract
Current clinical strategies to control the alloimmune response after transplantation do not fully prevent induction of the immunological processes which lead to acute and chronic immune-mediated graft rejection, and as such the survival of a solid organ allograft is limited. Experimental research on naturally occurring CD4+CD25highFoxP3+ Regulatory T cells (Tregs) has indicated their potential to establish stable long-term graft acceptance, with the promise of providing a more effective therapy for transplant recipients. Current approaches for clinical use are based on the infusion of freshly isolated or ex vivo polyclonally expanded Tregs into graft recipients with an aim to redress the in vivo balance of T effector cells to Tregs. However mounting evidence suggests that regulation of donor-specific immunity may be central to achieving immunological tolerance. Therefore, the next stages in optimizing translation of Tregs to organ transplantation will be through the refinement and development of donor alloantigen-specific Treg therapy. The altering kinetics and intensity of alloantigen presentation pathways and alloimmune priming following transplantation may indeed influence the specificity of the Treg required and the timing or frequency at which it needs to be administered. Here we review and discuss the relevance of antigen-specific regulation of alloreactivity by Tregs in experimental and clinical studies of tolerance and explore the concept of delivering an optimal Treg for the induction and maintenance phases of achieving transplantation tolerance.
Collapse
Affiliation(s)
- Pervinder Sagoo
- Department Transplantation, Immunoregulation and Mucosal Biology, MRC Centre for Transplantation, King's College London London, UK
| | | | | |
Collapse
|
12
|
Denecke C, Reutzel-Selke A, Sawitzki B, Boenisch O, Khalpey Z, Seifert M, Pratschke J, Volk HD, Tullius SG. Low-dose cyclosporine mediates donor hyporesponsiveness in a fully mismatched rat kidney transplant model. Transpl Immunol 2012; 26:176-85. [PMID: 22414756 DOI: 10.1016/j.trim.2012.02.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Revised: 02/23/2012] [Accepted: 02/27/2012] [Indexed: 12/11/2022]
Abstract
Tolerance induction protocols have been successfully tested in animal models, yet their compatibility with immunosuppressive drugs remains to be fully elucidated. Our own previous data have indicated that cyclosporine A (CsA) affects the balance of effector and regulatory mechanisms with low-dose CsA doses promoting hyporesponsiveness. Here, we present a fully mismatched rat kidney model in which low-dose CsA treatment induces donor hyporesponsiveness to secondary renal allografts. Lewis recipients of DA kidney grafts received low, medium or high doses of CsA × 10 days. By 30 days, the primary transplant was removed and a second transplant of donor origin was engrafted. Following low-dose CsA, but not high-dose CsA treatment of the primary recipient, secondary transplants were accepted long-term in the absence of immunosuppression. Regulatory T-cell function was unimpaired and independent of the CyA dosage. Of note, low-dose CsA significantly reduced alloantibody titers in primary recipients. Adoptive transfer of graft infiltrating cells or splenocytes from hyporesponsive recipients supported long-term acceptance of donor kidney allografts. These results demonstrate a dose-dependent and transferable "pro-tolerogenic" effect of low-dose CsA treatment. This model is of clinical relevance to test the interference of CsA with tolerance induction in the absence of additional immunosuppression.
Collapse
Affiliation(s)
- Christian Denecke
- Department of Visceral, Transplantation and Thoracic Surgery, Medizinische Universitaet Innsbruck, Austria; Division of Transplant Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Hoffen E, Polen E, Robertus-Teunissen M, Jonge N, Lahpor J, Gmelig-Meyling F, Weger R. High frequency of IL-4 producing helper T lymphocytes associated with a reduced incidence of heart allograft rejection. Transpl Int 2011. [DOI: 10.1111/j.1432-2277.2000.tb02023.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
14
|
Brown K, Badar A, Sunassee K, Fernandes MA, Shariff H, Jurcevic S, Blower PJ, Sacks SH, Mullen GED, Wong W. SPECT/CT lymphoscintigraphy of heterotopic cardiac grafts reveals novel sites of lymphatic drainage and T cell priming. Am J Transplant 2011; 11:225-34. [PMID: 21219574 PMCID: PMC6211618 DOI: 10.1111/j.1600-6143.2010.03388.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The normal function of lymphatic vessels is to facilitate the trafficking of antigen presenting cells to draining lymph nodes where they evoke an immune response. Donor lymphatic vessels are not connected to that of recipients' during organ transplantation. The pathophysiology of this disruption has received little attention. Murine heterotopic cardiac transplantation has been used extensively in transplantation research. Following vascularized organ transplantation, the main site of allosensitization is thought to be in the spleen of the recipient as a result of migration of donor passenger leukocytes via blood. Here, using Single Photon Emission Computed Tomography/Computerized Tomography (SPECT/CT) lymphoscintigraphy, we studied the pattern of lymphatic flow from mouse heterotopic abdominal cardiac grafts and identified mediastinal lymph nodes as the draining nodes for the donor graft. Staining with HY tetramer after transplantation of HY mismatched heart grafts and ELISPOT following allogeneic grafts to detect donor specific T cells revealed them as important sites for allosensitization. Our data indicates that mediastinal lymph nodes play a crucial role in the alloimmune response in this model, and should be used for ex vivo and adoptive transfer studies after transplantation in addition to the spleen.
Collapse
Affiliation(s)
- K. Brown
- MRC Centre for Transplantation, King’s College London, School of Medicine at Guy’s, King’s and St. Thomas’ Hospitals, London, UK
| | - A. Badar
- Division of Imaging Sciences, King’s College London, School of Medicine at Guy’s, King’s and St. Thomas’ Hospitals, London, UK
| | - K. Sunassee
- Division of Imaging Sciences, King’s College London, School of Medicine at Guy’s, King’s and St. Thomas’ Hospitals, London, UK
| | - M. A. Fernandes
- MRC Centre for Transplantation, King’s College London, School of Medicine at Guy’s, King’s and St. Thomas’ Hospitals, London, UK
| | - H. Shariff
- MRC Centre for Transplantation, King’s College London, School of Medicine at Guy’s, King’s and St. Thomas’ Hospitals, London, UK
| | - S. Jurcevic
- MRC Centre for Transplantation, King’s College London, School of Medicine at Guy’s, King’s and St. Thomas’ Hospitals, London, UK
| | - P. J. Blower
- Division of Imaging Sciences, King’s College London, School of Medicine at Guy’s, King’s and St. Thomas’ Hospitals, London, UK
| | - S. H. Sacks
- MRC Centre for Transplantation, King’s College London, School of Medicine at Guy’s, King’s and St. Thomas’ Hospitals, London, UK
| | - G. E. D. Mullen
- Division of Imaging Sciences, King’s College London, School of Medicine at Guy’s, King’s and St. Thomas’ Hospitals, London, UK
| | - W. Wong
- MRC Centre for Transplantation, King’s College London, School of Medicine at Guy’s, King’s and St. Thomas’ Hospitals, London, UK
| |
Collapse
|
15
|
Park J, Gao W, Whiston R, Strom TB, Metcalfe S, Fahmy TM. Modulation of CD4+ T lymphocyte lineage outcomes with targeted, nanoparticle-mediated cytokine delivery. Mol Pharm 2010; 8:143-52. [PMID: 20977190 DOI: 10.1021/mp100203a] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Within the immune system there is an exquisite ability to discriminate between "self" and "non-self" that is orchestrated by antigen-specific T lymphocytes. Genomic plasticity enables differentiation of naive CD4+ T lymphocytes into either regulatory cells (Treg) that express the transcription factor Foxp3 and actively prevent autoimmune self-destruction or effector cells (Teff) that attack and destroy their cognate target. An example of such plasticity is our recent discovery that leukemia inhibitory factor (LIF) supports Treg maturation in contrast to IL-6, which drives development of the pathogenic Th17 effector phenotype. This has revealed a LIF/IL6 axis in T cell development which can be exploited for modulation using targeted cytokine delivery. Here we demonstrate that LIF-loaded nanoparticles (NPs) directed to CD4+ T cells (i) oppose IL6-driven Th17 development; (ii) prolong survival of vascularized heart grafts in mice; and (iii) expand FOXP3+ CD4+ T cell numbers in a non-human primate model in vitro. In contrast, IL-6 loaded nanoparticles directed to CD4+ T cells increase Th17 development. Notably, nanoparticle-mediated delivery was demonstrated to be critical: unloaded nanoparticles and soluble LIF or IL-6 controls failed to recapitulate the efficacy of cytokine-loaded nanoparticles in induction and/or expansion of Foxp3+ cells or Th17 cells. Thus, this targeted nanoparticle approach is able to harness endogenous immune-regulatory pathways, providing a powerful new method to modulating T cell developmental plasticity in immune-mediated disease indications.
Collapse
Affiliation(s)
- Jason Park
- Department of Biomedical Engineering, Yale University, Malone Engineering Center, New Haven, Connecticut 06511, USA
| | | | | | | | | | | |
Collapse
|
16
|
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: 4.0] [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
|
17
|
Thompson LH, Whiston RA, Rakhimov Y, Taccioli C, Liu CG, Croce C, Metcalfe SM. A LIF/Nanog axis is revealed in T lymphocytes that lack MARCH-7, a RINGv E3 ligase that regulates the LIF-receptor. Cell Cycle 2010; 9:4213-21. [PMID: 20962578 PMCID: PMC3055204 DOI: 10.4161/cc.9.20.13543] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2010] [Accepted: 09/04/2010] [Indexed: 12/22/2022] Open
Abstract
Nanog is a stem cell transcription factor required for self-renewal and for maintaining pluripotency, and Nanog itself is regulated at least in part by leukaemia inhibitory factor (LIF)--a pluripotent cytokine of the IL6 family. MARCH-7 is an E-3 ligase linked to regulation of the LIF-receptor in T lymphocytes and T cells from mice that lack expression of MARCH-7 are hyper-responsive to activation signals and show a five-fold increase in LIF activity. Here we ask, does MARCH-7 influence the expression profile of Nanog during the synchronized entry of T cells into the cell cycle? We discovered that lack of MARCH-7 was permissive for Nanog expression at both transcript and protein levels during G₁/S: moreover, addition of exogenous LIF to the MARCH-7 null cells caused a further 13-fold induction of Nanog; other measured transcripts including TGFβ, p53 and STAT3 were relatively unchanged. Since lack of MARCH-7 altered responsiveness to activation signals we sought evidence for pre-existing regulatory miR's that might correlate with MARCH-7 gene dose using head-to-head comparisons between MARCH-7 null, heterozygous and wt spleen cells. 34 miRs were found including miR-346 that is known to target LIF transcripts and miR-346 is one of 16 miRs differentially expressed between hESCs and induced hiPSCs. Of the 34 miRs, 12 were known to be temporally regulated in embryonic nerve cells. In summary, in the absence of MARCH-7 a new signaling pathway is unmasked that involves Nanog expression in the T cell lineage. This is the first demonstration that T cells retain responsiveness to a LIF/Nanog axis and that this axis is linked to MARCH-7.
Collapse
|
18
|
Raimondi G, Sumpter TL, Matta BM, Pillai M, Corbitt N, Vodovotz Y, Wang Z, Thomson AW. Mammalian target of rapamycin inhibition and alloantigen-specific regulatory T cells synergize to promote long-term graft survival in immunocompetent recipients. THE JOURNAL OF IMMUNOLOGY 2009; 184:624-36. [PMID: 20007530 DOI: 10.4049/jimmunol.0900936] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Minimization of immunosuppression and donor-specific tolerance to MHC-mismatched organ grafts are important clinical goals. The therapeutic potential of regulatory T cells (Tregs) has been demonstrated, but conditions for optimizing their in vivo function posttransplant in nonlymphocyte-depleted hosts remain undefined. In this study, we address mechanisms through which inhibition of the mammalian target of rapamycin (Rapa) synergizes with alloantigen-specific Treg (AAsTreg) to permit long-term, donor-specific heart graft survival in immunocompetent hosts. Crucially, immature allogeneic dendritic cells allowed AAsTreg selection in vitro, with minimal expansion of unwanted (Th17) cells. The rendered Treg potently inhibited T cell proliferation in an Ag-specific manner. However, these AAsTreg remained unable to control T cells stimulated by allogeneic mature dendritic cells, a phenomenon dependent on the release of proinflammatory cytokines. In vivo, Rapa administration reduced danger-associated IL-6 production, T cell proliferation, and graft infiltration. Based on these observations, AAsTreg were administered posttransplant (day 7) in combination with a short course of Rapa and rendered >80% long-term (>150 d) graft survival, a result superior to that achieved with polyclonal Treg. Moreover, graft protection was alloantigen-specific. Significantly, long-term graft survival was associated with alloreactive T cell anergy. These findings delineate combination of transient mammalian target of Rapa inhibition with appropriate AAsTreg selection as an effective approach to promote long-term organ graft survival.
Collapse
Affiliation(s)
- Giorgio Raimondi
- Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| | | | | | | | | | | | | | | |
Collapse
|
19
|
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
|
20
|
Gao W, Thompson L, Zhou Q, Putheti P, Fahmy TM, Strom TB, Metcalfe SM. Treg versus Th17 lymphocyte lineages are cross-regulated by LIF versus IL-6. Cell Cycle 2009; 8:1444-50. [PMID: 19342884 PMCID: PMC2881570 DOI: 10.4161/cc.8.9.8348] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Within the immune system there is an exquisite ability to discriminate between "self" and "non-self" that is orchestrated by T lymphocytes. Discriminatory pathways guide differentiation of these lymphocytes into either regulatory (Treg) or effector (Teff) T cells, influenced by cues from the naïve T cell's immediate micro-environment as it responds to cognate antigen. Reciprocal pathways may lead to commitment of naïve T cells into either the protective tolerance-promoting Treg, or to the pro-inflammatory Th17 effector phenotype. Primary activation of CD4(+) lymphocytes stimulates their release of leukemia inhibitory factor (LIF), and Treg continue to release LIF in response to antigen, implying a role for LIF in tolerance. In contrast, interleukin- 6 (IL-6), although very closely related to LIF, promotes maturation of Th17 cells. Here we show that LIF and IL-6 behave as polar opposites in promoting commitment to the Treg and Th17 lineages. Unlike IL6, LIF supported expression of Foxp3, the Treg lineage transcription factor, and LIF opposed IL6 by suppressing IL-6-induced IL-17A protein release. In striking contrast, we found that IL6 effectively inhibited LIF signalling, repressing transcription of the LIF receptor gp190, and strongly inducing axotrophin/MARCH-7, a novel E3 ubitquitin ligase that we discovered to be active in degradation of gp190 protein. In vivo, anti-LIF treatment reduced donor-specific Treg in recipients of foreign spleen cells. Conversely, a single dose of biodegradable LIF nanoparticles, targeted to CD4, successfully manipulated the LIF/IL6 axis towards development of donor-specific Foxp3(+) Treg. The implications for therapy are profound, harnessing endogenous immune regulation by paracrine delivery of LIF to CD4(+) cells in vivo.
Collapse
Affiliation(s)
- Wenda Gao
- The Transplant Institute; Beth Israel Deaconess Medical Center and Harvard Medical School; Boston, MA USA
| | | | - Qiang Zhou
- The Transplant Institute; Beth Israel Deaconess Medical Center and Harvard Medical School; Boston, MA USA
| | - Prabhakar Putheti
- The Transplant Institute; Beth Israel Deaconess Medical Center and Harvard Medical School; Boston, MA USA
| | - Tarek M. Fahmy
- Department of Biomedical and Chemical Engineering; Yale University; New Haven, CT USA
| | - Terry B. Strom
- The Transplant Institute; Beth Israel Deaconess Medical Center and Harvard Medical School; Boston, MA USA
| | - Su M. Metcalfe
- Department of Surgery; University of Cambridge; Cambridge, UK
| |
Collapse
|
21
|
Horner BM, Ferguson KK, Randolph MA, Spencer JA, Carlson AL, Hirsh EL, Lin CP, Butler PEM. In vivo observations of cell trafficking in allotransplanted vascularized skin flaps and conventional skin grafts. J Plast Reconstr Aesthet Surg 2009; 63:711-9. [PMID: 19324598 DOI: 10.1016/j.bjps.2009.01.036] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2008] [Revised: 01/08/2009] [Accepted: 01/12/2009] [Indexed: 11/28/2022]
Abstract
The problem of allogeneic skin rejection is a major limitation to more widespread application of clinical composite tissue allotransplantation (CTA). Previous research examining skin rejection has mainly studied rejection of conventional skin grafts (CSG) using standard histological techniques. The aim of this study was to objectively assess if there were differences in the immune response to CSG and primarily vascularized skin in composite tissue allotransplants (SCTT) using in vivo techniques in order to gain new insights in to the immune response to skin allotransplants. CSG and SCTT were transplanted from standard Lewis (LEW) ad Wistar Furth (WF) to recipient transgenic green fluorescent Lewis rats (LEW-GFP). In vivo confocal microscopy was used to observe cell trafficking within skin of the transplants. In addition, immunohistochemical staining was performed on skin biopsies to reveal possible expression of class II major histocompatibility antigens. A difference was observed in the immune response to SCTT compared to CSG. SCTT had a greater density cellular infiltrate than CSG (p<0.03) that was focused more at the center of the transplant (p<0.05) than at the edges, likely due to the immediate vascularization of the skin. Recipient dendritic cells were only observed in rejecting SCTT, not CSG. Furthermore, dermal endothelial class II MHC expression was only observed in allogeneic SCTT. The immune response in both SCTT and CSG was focused on targets in the dermis, with infiltrating cells clustering around hair follicles (CSG and SCTT; p<0.01) and blood vessels (SCTT; p<0.01) in allogeneic transplants. This study suggests that there are significant differences between rejection of SCTT and CSG that may limit the relevance of much of the historical data on skin graft rejection when applied to composite tissue allotransplantation. Furthermore, the use of novel in vivo techniques identified characteristics of the immune response to allograft skin not previously described, which may be useful in directing future approaches to overcoming allograft skin rejection.
Collapse
Affiliation(s)
- Benjamin M Horner
- Plastic Surgery Research Laboratory, Department of Surgery, WAC 435, Massachusetts General Hospital, Boston, MA 02114, USA
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Francuski M, Reutzel-Selke A, Weiss S, Pascher A, Jurisch A, Ulrich F, Schumacher G, Faber W, Kohler S, Volk HD, Neuhaus P, Tullius SG, Pratschke J. Donor brain death significantly interferes with tolerance induction protocols. Transpl Int 2008; 22:482-93. [PMID: 18954365 DOI: 10.1111/j.1432-2277.2008.00776.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Studies in rodents showed that antibodies are able to induce tolerance of allografts. As clinical results are unsatisfactory and deceased donors are still the main source of organ transplants, we investigated whether donor brain-death impacts on tolerance induction after experimental kidney transplantation. Anti-CD4 monoclonal antibodies (RIB 5/2; 2.5 mg/kg x 5 days) treated and untreated recipients of brain-dead donor grafts were compared with RIB 5/2 treated and untreated recipients of living donor grafts (F344-to-Lewis). All recipients received low-dose CsA (1.5 mg/kg x 10 days). Kidneys were recovered 4, 16 and 40 weeks after transplantation and examined by morphology, immunohistology and flow cytometry. Renal function was monitored monthly. RIB 5/2 treatment significantly decreased proteinuria in recipients of living donor allografts when compared with living donor controls. After 40 weeks, inflammatory cell infiltration and MHC class II expression were reduced while morphologic alterations were minimal. In contrast, treatment of brain-dead graft recipients had no impact on graft function. Structural changes and graft infiltration were comparable to brain-dead donor controls at all time points. RIB 5/2 treatment significantly improved graft function in recipients of living donor grafts; however, it was not effective in recipients of brain-dead donor organs.
Collapse
Affiliation(s)
- Maja Francuski
- Department of General, Visceral, and Transplantation Surgery, Charité, Campus Virchow Clinic, Universitätsmedizin Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Regulation and Privilege in Transplantation Tolerance. J Clin Immunol 2008; 28:716-25. [DOI: 10.1007/s10875-008-9249-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2008] [Accepted: 08/01/2008] [Indexed: 02/03/2023]
|
24
|
Role of persistence of antigen and indirect recognition in the maintenance of tolerance to renal allografts. Transplantation 2008; 85:270-80. [PMID: 18212633 DOI: 10.1097/tp.0b013e31815e8eed] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND We have previously shown that a 12-day treatment with cyclosporine A (CyA) facilitates induction of tolerance to class-I disparate kidneys, as demonstrated by acceptance of second, donor-matched kidneys without immunosuppression. In the present study, we have examined 1) the duration of tolerance in the absence of donor antigen and 2) the pathway of antigen recognition determining maintenance or loss of tolerance. METHODS Seventeen miniature swine received class-I mismatched kidneys with 12 days of CyA, and received second donor-matched kidneys without immunosuppression at 0, 1, 3, or 4 months after nephrectomy of the primary graft. Five were sensitized 6 weeks after nephrectomy of the primary graft, three with donor-matched skin grafts, and two with donor class-I peptides to eliminate direct pathway involvement. In addition, two long-term tolerant animals received class-I peptides. RESULTS Rejection of second grafts required at least a 3 month absence of donor antigen. Although donor-matched skin grafts in animals tolerant to kidneys induced antidonor cytotoxic T lymphocyte responses, second renal transplants revealed no evidence of sensitization. In contrast, immunization of recipients with donor class-I peptides after nephrectomy of the primary graft led to loss of tolerance at both T-cell and B-cell levels, as evidenced by rejection of the second graft in 5 days and development of antidonor immunoglobulin G. Peptide immunization of long-term tolerant in recipients bearing long-term renal grafts did not break tolerance. CONCLUSIONS These data indicate that the renal allograft is required for the indefinite maintenance of tolerance, that indirect antigen presentation is capable of breaking tolerance, and that in tolerant animals, direct antigen presentation may suppress rejection, allowing tolerance to persist.
Collapse
|
25
|
Muthukumarana PADS, Zheng XX, Rosengard BR, Strom TB, Metcalfe SM. In primed allo-tolerance, TIM-3-Ig rapidly suppresses TGFbeta, but has no immediate effect on Foxp3. Transpl Int 2008; 21:593-7. [PMID: 18282245 DOI: 10.1111/j.1432-2277.2008.00654.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
T-cell immunoglobulin mucin-3 (TIM-3) is only expressed by differentiated TH1 cells following their proliferative response to antigen, functioning to terminate TH1-mediated immunity upon binding to the TIM-3 ligand, galectin-9. This critical regulatory process involves Treg cells via their stable expression of galectin-9. Soluble TIM-3-Ig blocks galectin-9 and prevents induction of peripheral tolerance. Here we have looked for evidence that TIM-3-Ig might also break established regulatory tolerance. Using allo-primed spleen cells cultured ex vivo and challenged with irradiated donor-type stimulator cells either alone or together with 20 microg/ml TIM-3-Ig, we measured daily cytokine release [IL2, inferon gamma (INFgamma), transforming growth factor beta (TGFbeta), IL6, IL10] and cellular Foxp3 protein. In allo-tolerance, a specific effect of TIM-3-Ig was some fourfold reduction in TGFbeta. Foxp3 was induced in the allo-tolerant response to donor and this was not altered by TIM-3-Ig over the 5-day culture period. No Foxp3 was detected in either rejected or donor stimulator cells at any time. Thus, in an ex vivo model of in vivo tolerance to heart allografts, TIM-3-Ig therapy appears to reduce the stable tolerogenic environment by a rapid and specific repression of TGFbeta release.
Collapse
|
26
|
Hara M, Chosa E, Onitsuka T. The spleen's role in transplantation immunology. Transpl Immunol 2008; 18:324-9. [DOI: 10.1016/j.trim.2007.09.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2007] [Revised: 09/09/2007] [Accepted: 09/17/2007] [Indexed: 11/29/2022]
|
27
|
del Rio ML, Pabst O, Ramirez P, Penuelas-Rivas G, Förster R, Rodriguez-Barbosa JI. The thymus is required for the ability of FTY720 to prolong skin allograft survival across different histocompatibility MHC barriers. Transpl Int 2007; 20:895-903. [PMID: 17854447 DOI: 10.1111/j.1432-2277.2007.00539.x] [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] [Indexed: 01/22/2023]
Abstract
The immunosuppressive effect of FTY720 is associated with the reversible sequestration of lymphocytes from the blood and the spleen into secondary lymphoid organs and reduced egress of mature thymocytes from the thymus. This work was designed to dissect the differential effect of FTY720 on CD4 and CD8 T cell-mediated mechanisms of skin graft rejection in the presence (euthymic) or absence (thymectomized) of thymic output. To that end, untreated and FTY720-treated euthymic (Euthy) and thymectomized (ATX) mice received skin allografts across a full, class II or class I major histocompatibility complex (MHC) mismatched (MM) barriers and graft survival was monitored. We demonstrate that a short course of FTY720 treatment significantly augments the survival of full, class I and class II MHC MM skin grafts compared to the nontreated controls. Interestingly, FTY720-treated Euthy recipients showed a significantly prolonged skin allograft survival compared to FTY720-treated ATX mice. These results together show that FTY720 impairs both CD4 and CD8 T cell-mediated mechanisms of rejection and, more importantly, the presence of the thymus is necessary for the ability of FTY720 to modulate skin allograft rejection across different histocompatibility MHC barriers.
Collapse
Affiliation(s)
- María L del Rio
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | | | | | | | | | | |
Collapse
|
28
|
Abstract
In 1943, Gibson and Medawar opened the modern era of transplantation research with a paper on the problem of skin allograft rejection. Ten years later Billingham, Brent and Medawar demonstrated that it was possible to induce selective immune acceptance of skin grafts in mice, a state of tolerance. After over six decades, however, the precise mechanism of skin allograft rejection remains still ill-defined. Furthermore, it has not been possible to achieve reliably clinical tolerance allowing the widespread application of skin allotransplantation techniques. The first successful applications of skin allotransplantation have included the hand and face. However, complications from the chronic immunosuppression regimens limit the application of these techniques. Induction of tolerance to skin (and the other tissues in the allograft) would be the most effective way to overcome all these difficulties, but this is yet to be achieved reliably, stimulating some to look for other ways to surmount the current limitations. This paper summarizes alternatives to enlarge the scope of skin allotransplantation techniques, current understanding of mechanisms of skin rejection, and the utility and limitations of animal models used to study skin rejection and tolerance induction. Finally, manipulation strategies to achieve skin tolerance are outlined.
Collapse
Affiliation(s)
- Benjamin M Horner
- Transplantation Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02129, USA
| | | | | | | |
Collapse
|
29
|
Muthukumarana P, Chae WJ, Maher S, Rosengard BR, Bothwell ALM, Metcalfe SM. Regulatory transplantation tolerance and "stemness": evidence that Foxp3 may play a regulatory role in SOCS-3 gene transcription. Transplantation 2007; 84:S6-11. [PMID: 17632414 DOI: 10.1097/01.tp.0000269116.06510.db] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Immune self-tolerance is controlled by a subset of T lymphocytes that are regulatory (Treg) and epigenetically programmed to suppress autoreactive immune effector cells in vivo. Treg require expression of Foxp3, a transcription factor that not only represses the interleukin-2 gene promoter, but also sequesters key mediators of T-cell signal transduction by complexing with cytoplasmic NFAT and NFkappaB. We have discovered that expression of Foxp3 is linked to two stem cell-related factors, namely leukemia inhibitory factor (LIF) and axotrophin. Because both LIF and axotrophin each influence Foxp3, we now ask if reciprocal cross-talk occurs; for example, does Foxp3 in turn influence LIF and/or axotrophin? We compared the effect of wt-Foxp3 versus mutant DeltaE251-Foxp3, which lacks transcriptional activity, on transcript levels of axotrophin, LIF, and suppressor of cytokine signaling-3 (SOCS-3; a feedback inhibitor of LIF) in the Jurkat human T-cell line. Unexpectedly, a 50-fold increase in SOCS-3 transcripts occurred in the DeltaE251-Foxp3 cells, coincident with a dramatic decrease in LIF transcription. This implies that, either directly or indirectly, transcription of SOCS-3 is negatively regulated by wt-Foxp3. Suppression of SOCS-3 by Foxp3 would support a model wherein Foxp3 promotes LIF signaling in Treg and is further evidence of reciprocity between Foxp3, LIF, and axotrophin.
Collapse
Affiliation(s)
- Poorni Muthukumarana
- Department of Surgery, University of Cambridge, Addenbrooke's Hospital, Hills Road, Cambridge, United Kingdom
| | | | | | | | | | | |
Collapse
|
30
|
Pu LY, Wang XH, Zhang F, Li XC, Yao AH, Yu Y, Lv L, Li GQ. Adoptive transfusion of ex vivo donor alloantigen-stimulated CD4(+)CD25(+) regulatory T cells ameliorates rejection of DA-to-Lewis rat liver transplantation. Surgery 2007; 142:67-73. [PMID: 17630002 DOI: 10.1016/j.surg.2007.02.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2006] [Revised: 02/15/2007] [Accepted: 02/24/2007] [Indexed: 01/27/2023]
Abstract
BACKGROUND Adoptive transfusion of splenocytes from long-term survivors of a tolerance model of rat orthotopic liver transplantation can induce acceptance of liver allografts in a rejection model preconditioned with donor gamma-irradiation before liver transplantation. Recent studies suggest that the regulatory T cells (Treg cells) in splenocytes from long-term survivors play an important role in the induction of liver graft tolerance, but this observation was made from a rejection model preconditioned with donor gamma-irradiation; little is known about the role of Treg cells in liver graft rejection using a naive rejection model. In this study, we examined the therapeutic potential of CD4(+)CD25(+) Treg cells in a naive rejection model of rat liver transplantation. METHODS Freshly isolated or ex vivo alloantigen-stimulated CD4(+)CD25(+) Treg cells (1 x 10(6) cells) from naive Lewis RT(1) (LEW) rats were adoptively transferred into another LEW rat on days 1 and 7 after liver transplantation from a Dark Agouti RT1(a) (DA) rat. Recipients were treated with or without oral tacrolimus (FK506) (0.1 mg/kg/day) from days 1 to 7 after transplantation. For ex vivo alloantigen-stimulation, CD4(+)CD25(+) Treg cells from LEW rats were cocultured with mitomycin C-treated DA (donor alloantigen specific) or Brown Norway (BN)(RT1(n), third party) splenocytes for 72 hours. Ex vivo alloantigen-specific CD4(+)CD25(-) T-cell proliferation responses were assessed with fresh and stimulated CD4(+)CD25(+) Treg cells. RESULTS Freshly isolated, donor alloantigen-stimulated and third-party alloantigen- stimulated CD4(+)CD25(+) Treg cells suppressed antigen-specific CD4(+)CD25(-) T-cell proliferation ex vivo, and adoptive transfusion of these 3 kinds of CD4(+)CD25(+) Treg cells prolonged survival of the liver allografts. The group transfused with the donor alloantigen-stimulated CD4(+)CD25(+) Treg cells had the greatest mean survival among the 3 groups (fresh Treg cells, 21 +/- 2 days, n = 6; third-party alloantigen-stimulated Treg cells, 20 +/- 2 days, n = 6; donor alloantigen-stimulated Treg cells, 30 +/- 2 days, n = 6). When combined with short-term tacrolimus administration, adoptive transfusion of donor antigen-stimulated Treg cells induced the greatest survival time in recipients (greater than 60 days; n = 6). CONCLUSION Adoptive transfusion of ex vivo donor alloantigen-stimulated CD4(+)CD25(+) Treg cells combined with short-term tacrolimus treatment may represent a new strategy for preventing rejection after liver transplantation.
Collapse
Affiliation(s)
- Li-Yong Pu
- Liver Transplantation Center, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Hall BM, Plain KM, Verma ND, Tran GT, Boyd R, Robinson CM, Nicolls MR, Berger ME, Nomura M, Hodgkinson SJ. Transfer of Allograft Specific Tolerance Requires CD4+CD25+T Cells but Not Interleukin-4 or Transforming Growth Factor–β and Cannot Induce Tolerance to Linked Antigens. Transplantation 2007; 83:1075-84. [PMID: 17452898 DOI: 10.1097/01.tp.0000259553.66185.2f] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND The mechanisms by which CD4+T cells, especially CD4+ CD25+T cells, transfer allograft specific tolerance are poorly defined. The role of cytokines and the effect on antigen-presenting cells is not resolved. METHODS Anti-CD3 monoclonal antibody (mAb) therapy induced tolerance to PVG heterotopic cardiac transplantation in DA rats. Peripheral CD4+T cells or CD4+ CD25+ and CD4+ CD25-T cell subsets were adoptively transferred to irradiated DA hosts grafted with PVG heart grafts. For specificity studies, tolerant CD4+T cells were transferred to hosts with Lewis or (PVGxLewis)F1 heart grafts. Cytokine mRNA induction and the requirement for interleukin (IL)-4 and transforming growth factor (TGF)-beta in the transfer of tolerance was assessed. RESULTS CD4+T cells transferred specific tolerance and suppressed naïve CD4+T cells capacity to effect rejection of PVG but not Lewis grafts. (PVGxLewis)F1 grafts had a major rejection episode but recovered. Later these hosts accepted PVG but not Lewis skin grafts. Adoptive hosts restored with tolerant or naïve cells had similar levels of mRNA expression for all Th1 and Th2 cytokines and effector molecules assayed. Transfer of tolerance by CD4+T cells was not blocked by mAb to IL-4 or TGF-beta. CD4+ CD25-T cells from either naïve or tolerant hosts effected rejection. In contrast neither tolerant nor naïve CD4+ CD25+T cells restored rejection. CONCLUSIONS Specific tolerance transfer required CD4+ containing CD4+ CD25+T cells. An inflammatory response with induction of mRNA for Th1 and Th2 cytokines plus cytotoxic effector molecules occurred, but IL-4 and TGF-beta were not essential. Inhibition of antigen presenting cells was not the sole mechanism as there was no linked tolerance.
Collapse
Affiliation(s)
- Bruce M Hall
- Immune Tolerance Laboratory, University of New South Wales, Australian Technology Park, New South Wales, Australia.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Bushell A, Wood K. GITR ligation blocks allograft protection by induced CD25+CD4+ regulatory T cells without enhancing effector T-cell function. Am J Transplant 2007; 7:759-68. [PMID: 17391121 DOI: 10.1111/j.1600-6143.2006.01716.x] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The induction of operational tolerance prior to transplant could provide a solution to the complications of current immunosuppression in transplantation. In rodents, operational tolerance frequently correlates with the presence of CD25(+)CD4(+) regulatory T cells (Tregs) but their function is usually demonstrated by adoptive transfer into lymphopenic hosts leading some to question their relevance to normal immunocompetent recipients. The role of these cells in primary transplant recipients has been explored using anti-CD25 antibody but specific targeting of Treg is not possible since CD25 is also up-regulated on activated effector T cells. To overcome this limitation we targeted the Treg associated molecule GITR in tolerized primary transplant recipients. This reverses regulation resulting in acute allograft rejection. This is not due to co-stimulation of effector cells since rejection mediated by isolated populations of CD4(+)CD25(-) or CD8(+)CD25(-) T cells transferred into Rag(-/-) mice was not enhanced by anti-GITR antibody. Furthermore, GITR cross-linking does not provide co-stimulation for in vitro proliferation of the same CD4(+)CD25(-) or CD8(+)CD25(-) T-cell populations in response donor-strain APC. Thus, CD4(+)CD25(+)GITR(+) Treg play an essential role in early graft protection in primary transplant recipients following tolerance induction providing further support for protocols that might generate similar populations in clinical transplantation.
Collapse
Affiliation(s)
- A Bushell
- Nuffield Department of Surgery, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK.
| | | |
Collapse
|
33
|
Abstract
The father of cardiac transplantation, Norman Shumway, famously predicted that tolerance was the future of the field, and always would be. Although his prediction remains true to date, significant progress has been made toward this goal, the "Holy Grail" for transplant clinicians. Current efforts are fueled by disappointing long-term outcomes associated with chronic immunosuppression, and the promise that partial or complete tolerance will impact long-term results favorably. This article provides a clinical definition of tolerance primarily based on lessons learned from animal heart allograft models. It reviews several promising strategies for inducing tolerance and detecting its presence through the use of biomarkers in peripheral blood or the graft, and outlines a possible path toward making this vision a clinical reality.
Collapse
Affiliation(s)
- Richard N Pierson
- Baltimore VA Medical Center, University of Maryland Medical School, Baltimore, MD 21201, USA.
| |
Collapse
|
34
|
Abstract
Monoclonal antibodies (MAb) have been shown to be effective in inducing immune tolerance in transplantation and autoimmunity. Several different MAb have tolerogenic properties and their effect has been studied in a range of experimental animal models and, in some cases, in clinical trials. The tolerant state seems to be maintained by CD4+ regulatory T cells (Treg), induced in the periphery, capable of suppressing other T cells specific for the same antigens or antigens presented by the same antigen presenting cells. Furthermore, following the initial induction of Treg cells under MAb treatment, Treg cells themselves can maintain the tolerant state in a dominant way in the absence of the therapeutic MAb or other immunosuppressive agents, and are able to recruit other T cells into the regulatory pool--a process named infectious tolerance.
Collapse
Affiliation(s)
- Ana Agua-Doce
- Instituto de Medicina Molecular, Universidade de Lisboa, Portugal
| | | |
Collapse
|
35
|
Muthukumarana PADS, Lyons GE, Miura Y, Thompson LH, Watson T, Green CJ, Shurey S, Hess AD, Rosengard BR, Metcalfe SM. Evidence for functional inter-relationships between FOXP3, leukaemia inhibitory factor, and axotrophin/MARCH-7 in transplantation tolerance. Int Immunopharmacol 2006; 6:1993-2001. [PMID: 17161353 DOI: 10.1016/j.intimp.2006.09.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2006] [Accepted: 09/15/2006] [Indexed: 12/15/2022]
Abstract
In an ex vivo mouse model, regulatory transplantation tolerance is not only linked to Foxp3, but also to release of leukaemia inhibitory factor (LIF) and to expression of axotrophin (also known as MARCH-7), a putative ubiquitin E3 ligase associated with feedback control of T cell activation and of T cell-derived LIF. Given this coordinate correlation with tolerance, we now ask if Foxp3 expression is influenced by LIF or by axotrophin. In spleen cells from allo-rejected mice we found that exogenous LIF reduced interferon gamma release in response to donor antigen by 50%, but LIF had no direct effect on levels of Foxp3 protein in allo-primed cells that were either tolerant, or aggressive, for donor antigen. However, we did find an effect of axotrophin on Foxp3: in the axotrophin null mouse, thymic Foxp3 transcripts were reduced compared to axotrophin wildtype littermates. To test whether these findings in the mouse were of potential significance in man we measured transcript levels of axotrophin and LIF in peripheral blood cell samples collected for a recently published clinical study concerning haematopoietic stem cell recipients. In controls, human peripheral blood CD4+CD25+cells contained significantly more FOXP3 and axotrophin than CD4+CD25-cells. In bone marrow autograft recipients, where peripheral blood cell samples directly represent both the grafted tissue and the immune response, both FOXP3 and axotrophin negatively correlated with graft versus host disease (GVHD). These data suggest that (i) thymic Foxp3+T cell development is influenced by axotrophin; and (ii) clinical auto-GVHD inversely correlates with axotrophin transcript expression as has been previously reported for FOXP3.
Collapse
|
36
|
Yong Z, Chang L, Mei YX, Yi L. Role and mechanisms of CD4+CD25+ regulatory T cells in the induction and maintenance of transplantation tolerance. Transpl Immunol 2006; 17:120-9. [PMID: 17306743 DOI: 10.1016/j.trim.2006.09.005] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2006] [Revised: 08/22/2006] [Accepted: 09/08/2006] [Indexed: 12/23/2022]
Abstract
To gain transplantation tolerance between donor organs and hosts is the ultimate goal of all sorts of organ transplantations. Induction of regulatory T cells has been demonstrated to lead to transplantation tolerance. This paper will review subsets of regulatory T cells, the role and mechanisms of CD4(+)CD25(+) regulatory T cells (Tregs) in graft rejection and tolerance, pathway used by Tregs to recognized alloantigens, pathways of Tregs homing into the graft and effects of immunosuppression on Tregs. It was well known that Tregs play a pivotal role in transplantation tolerance. The mechanisms by which Tregs exert their regulatory effect in the induction and maintenance of transplantation tolerance, anthropogenically, consist of physical cell-to-cell contact with potential target cells, autocrine and paracrine properties. ICAM-1, TGF-beta, CTLA-4, GITR and OX40 (CD134), etc. are involved in the regulatory function of Tregs through cell-to-cell contact mechanism. IL-10 and TGF-beta are two important soluble mediators involved in the autocrine mechanism by which Tregs exert their regulatory function. Paracrine properties refer to re-educate potentially destructive alloresponsive T cells to gain regulatory function. All that discussed above could illustrate, at least partially, how naturally occurring Tregs exert their regulatory function in vivo as they constitute only 5-10% of peripheral CD4(+) T cells.
Collapse
Affiliation(s)
- Zhang Yong
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Medical Collage, Xi'an Jiaotong University, Xi'an 710061, Shaanxi Province, China
| | | | | | | |
Collapse
|
37
|
Cobbold SP, Adams E, Graca L, Daley S, Yates S, Paterson A, Robertson NJ, Nolan KF, Fairchild PJ, Waldmann H. Immune privilege induced by regulatory T cells in transplantation tolerance. Immunol Rev 2006; 213:239-55. [PMID: 16972908 DOI: 10.1111/j.1600-065x.2006.00428.x] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Immune privilege was originally believed to be associated with particular organs, such as the testes, brain, the anterior chamber of the eye, and the placenta, which need to be protected from any excessive inflammatory activity. It is now becoming clear, however, that immune privilege can be acquired locally in many different tissues in response to inflammation, but particularly due to the action of regulatory T cells (Tregs) induced by the deliberate therapeutic manipulation of the immune system toward tolerance. In this review, we consider the interplay between Tregs, dendritic cells, and the graft itself and the resulting local protective mechanisms that are coordinated to maintain the tolerant state. We discuss how both anti-inflammatory cytokines and negative costimulatory interactions can elicit a number of interrelated mechanisms to regulate both T-cell and antigen-presenting cell activity, for example, by catabolism of the amino acids tryptophan and arginine and the induction of hemoxygenase and carbon monoxide. The induction of local immune privilege has implications for the design of therapeutic regimens and the monitoring of the tolerant status of patients being weaned off immunosuppression.
Collapse
Affiliation(s)
- Stephen P Cobbold
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Koenen HJPM, Joosten I. Antigen-Specific Regulatory T-Cell Subsets in Transplantation Tolerance. Hum Immunol 2006; 67:665-75. [PMID: 17002896 DOI: 10.1016/j.humimm.2006.05.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2006] [Accepted: 05/26/2006] [Indexed: 12/11/2022]
Abstract
Regulatory T cells (Treg) are critical controllers of the immune response. Disturbed Treg function results in autoimmunity, whereas in transplantation Treg are crucial in graft survival and transplant tolerance. Hence therapeutic modalities that influence Treg numbers or function hold great clinical opportunity. Ahead of us are clinical trails studying in vivo Treg induction protocols and immunotherapy with ex vivo expanded Treg. Here we discuss the preferential use and/or induction of antigen-specific Treg subsets with high suppressive power and migratory capacity as a potential therapeutic tool to prevent solid organ transplantation rejection. Accordingly, ex vivoselection procedures to induce and isolate highly suppressive antigen-specific Treg (subsets) are needed. This subject, as well as the Treg-facilitating potential of immunosuppressive agents, is discussed.
Collapse
Affiliation(s)
- Hans J P M Koenen
- Department of Blood Transfusion and Transplantation Immunology, Radboud University Medical Center Nijmegen, Nijmegen, The Netherlands.
| | | |
Collapse
|
39
|
Waldmann H, Adams E, Fairchild P, Cobbold S. Infectious tolerance and the long-term acceptance of transplanted tissue. Immunol Rev 2006; 212:301-13. [PMID: 16903922 DOI: 10.1111/j.0105-2896.2006.00406.x] [Citation(s) in RCA: 134] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Short courses of antibody treatment aimed at blocking the coreceptors CD4 and CD8 and/or costimulatory molecules such as CD40L are able to bring about long-term acceptance and tolerance of allogeneic transplants. This tolerant state is operational, in that potential effector cells remain but are tightly regulated through the induction of antigen-specific CD4+ regulatory T cells (Tregs). CD4+ CD25+ FoxP3+ Tregs appear to play a prominent role, although other categories of Tregs have been documented. Transforming growth factor beta (TGFbeta) has been found to play a major role in the induction of the tolerant state with therapeutic antibodies as well as promoting the induction of FoxP3+ T cells from naïve populations. The observation that Tregs can be found in tolerated grafts has led to the idea that they may interact with the grafted tissue to establish a state of acquired privilege symmetrical with a similar privileged microenvironment around antigen-presenting cells in lymphoid tissues. Dampening of aggressive immune responses by Tregs allows antigen to persist and be presented in an innocuous way to promote tolerance in new cohorts of T cells throughout the life of the tolerated graft. Regulation may operate at many stages of an immune response, even as a censor at the terminal differentiation stages of effector function.
Collapse
|
40
|
Albert MH, Anasetti C, Yu XZ. T regulatory cells as an immunotherapy for transplantation. Expert Opin Biol Ther 2006; 6:315-24. [PMID: 16548760 DOI: 10.1517/14712598.6.4.315] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Advances in immunosuppressive therapies have made tissue and organ transplantation a common procedure in clinical medicine. However, true donor and recipient tolerance is not regularly achieved and almost all transplant recipients continue to require immunosuppressants throughout life, which is associated with side effects of the drugs. The identification and characterisation of regulatory T cells (Tregs) has recently opened up exciting opportunities for new ways of adoptive immunotherapy in transplantation. CD4+CD25+ Tregs of thymic origin have been shown to be key regulators of unseasoned immune responses in mice and in humans, preventing graft-versus-host disease and organ graft rejection in the transplantation setting. Although these cells can be found in the peripheral blood of healthy individuals, their isolation to a satisfying degree of purity is time-consuming and ineffective. Therefore, a variety of different methods to expand or induce regulatory T cells ex vivo have been advocated. Antigen-specific activation of Tregs is a prerequisite for their optimal function, making the design of new strategies to create and expand antigen-specific Tregs highly desirable. This review will focus on recent advances achieved in the field of transplantation tolerance using naturally occurring Tregs (CD4+CD25+), as well as other Tregs, and will discuss future applications of these cells in immunotherapy.
Collapse
|
41
|
Negative costimulatory molecules: the proximal of regulatory T cells? Med Hypotheses 2006; 67:841-7. [PMID: 16762512 DOI: 10.1016/j.mehy.2006.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2006] [Accepted: 04/04/2006] [Indexed: 10/24/2022]
Abstract
Regulatory T cells (Tregs) are a central mechanism of immune regulation. It is well known that their regulatory effect is antigen-specific and depends on cell-cell contact. In addition, some immunological phenomenon such as linked suppression and iDC-induced tolerance are related with Tregs. But the surface markers, which reliably distinguish Treg from other T cell populations, and the regulatory mechanism still remain to be further revealed. Negative costimulatory molecule (NCM) family is one natural intrinsic mechanism that delivers the negative signal into cytoplasma to modulate immunoresponse and its expression can be induced not only on the immune cells but also on the parenchymal cells. Based on the present knowledge, we hypothesize NCMs are the specific surface markers to define Tregs. Tregs are one kind of activated T cells with high expression of NCM receptor and have the capability to induce NCM ligands expression on the membrane of APCs and the target cells of the activated cells. The NCM receptor-ligand complexes deliver negative signal into lymphocytes to regulate the immune response. This hypothesis remains to be fully elucidated.
Collapse
|
42
|
Neujahr DC, Chen C, Huang X, Markmann JF, Cobbold S, Waldmann H, Sayegh MH, Hancock WW, Turka LA. Accelerated Memory Cell Homeostasis during T Cell Depletion and Approaches to Overcome It. THE JOURNAL OF IMMUNOLOGY 2006; 176:4632-9. [PMID: 16585554 DOI: 10.4049/jimmunol.176.8.4632] [Citation(s) in RCA: 117] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Partial T cell depletion is used in solid organ transplantation as a valuable strategy of peritransplant induction immunosuppression. Using a murine cardiac allograft model, we recently demonstrated that this led to lymphopenia-induced (homeostatic) proliferation among the residual nondepleted lymphocytes. Rather than promoting tolerance, peritransplant T cell-depleting Abs actually resulted in resistance to tolerance induction by costimulatory blockade. In this study we show that memory T cells predominate shortly after subtotal lymphodepletion due to two distinct mechanisms: relative resistance to depletion and enhanced homeostatic proliferation. In contrast, regulatory cells (CD4+ CD25+ Foxp3+) are depleted as efficiently as nonregulatory cells and exhibit reduced homeostatic expansion compared with memory cells. The resistance to tolerance induction seen with subtotal T cell depletion can be overcome in two different ways: first, by the adoptive transfer of additional unprimed regulatory cells at the time of transplant, and second, by the adjunctive use of nondepleting anti-CD4 and anti-CD8 mAbs, which effectively block homeostatic expansion. We conclude that the resistance to tolerance induction seen after subtotal lymphocyte depletion can be attributed to alterations in the balance of naive, memory, and regulatory T cells. These data have clinically relevant implications related to the development of novel strategies to overcome resistance to tolerance.
Collapse
Affiliation(s)
- David C Neujahr
- Department of Medicine, University of Pennsylvania, Philadelphia 19104, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
Our ability to harness tolerance mechanisms will have a major impact in organ transplantation. It should enable drug minimization, and eventually, the elimination of all immunosuppressive drugs. An improved understanding of the biology of regulatory T cells will make it possible to replace current induction regimens with those favouring the selective vaccination of T cells that prevent graft rejection. Once regulation is established, the continued supply of graft antigens should empower T cell regulation to become the dominant natural mechanism to prevent graft rejection.
Collapse
Affiliation(s)
- Herman Waldmann
- Sir William Dunn School of Pathology, South Parks Road, Oxford OX1 3RE, UK.
| | | | | | | | | | | | | |
Collapse
|
44
|
Waldmann H, Graca L, Adams E, Fairchild P, Cobbold S. Regulatory T cells in transplantation tolerance. Curr Top Microbiol Immunol 2005; 293:249-64. [PMID: 15981483 DOI: 10.1007/3-540-27702-1_11] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Our ability to harness tolerance mechanisms will have a major impact in organ transplantation if it becomes possible to minimize drug maintenance, or even wean off immunosuppressive drugs. An improved understanding of the biology of regulatory T cells will make it possible to replace current induction regimens with those favouring the vaccination and selection of T cells that prevent graft rejection. Once tolerance is established, the continuous supply of graft antigens should sustain T cell mediated regulation as the dominant mechanism preventing graft rejection.
Collapse
Affiliation(s)
- H Waldmann
- Sir William Dunn School of Pathology, South Parks Road, Oxford OX1 3RE, UK.
| | | | | | | | | |
Collapse
|
45
|
Heslan JM, Beriou G, Le Luduec JB, Guillonneau C, Anegon I, Soulillou JP, Cuturi MC, Chiffoleau E. Accumulation of T Cells with Potent Regulatory Properties and Restricted Vβ7-TCR Rearrangements in Tolerated Allografts. Transplantation 2005; 80:1476-84. [PMID: 16340794 DOI: 10.1097/01.tp.0000185198.07663.ba] [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 We have previously demonstrated that a short-course treatment with LF15-0195, a 15-deoxyspergualin analogue, induces donor-specific tolerance of cardiac allografts in rats and expansion of splenic CD4CD25 regulatory T cells. METHODS To further characterize long-term tolerance in this model, we have analyzed the phenotype, regulatory properties and TCR-Vbeta usage of the T cells infiltrating the tolerated allografts. RESULTS We demonstrate that the tolerated allografts express high levels of FoxP3 transcripts and contain a large number of CD4 T cells, half of which express CD25. Moreover, T cells from these tolerated allografts are very powerful at transferring tolerance to a subsequent allograft recipient, demonstrating the presence of potent regulatory T cells at the site of the graft. Interestingly, the T cells infiltrating the tolerated allografts systematically display restricted Vbeta7 TCR rearrangements. CONCLUSION These results demonstrate in this model of tolerance, a specific accumulation of T cells with potent regulatory properties and exhibiting restricted Vbeta7-TCR rearrangements at the graft site.
Collapse
Affiliation(s)
- Jean-Marie Heslan
- Institut National de la Santé et de la Recherche Médicale Unité 643 (INSERM U643) and Institut de Transplantation Et de Recherche en Transplantation, CHU Hotel Dieu, Nantes, France
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Buer J, Westendorf AM, Zeng AP, He F, Hansen W, Probst-Kepper M. Mechanisms of Central and Peripheral T-Cell Tolerance: An Update. Transfus Med Hemother 2005. [DOI: 10.1159/000089128] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
47
|
Metcalfe SM. Axotrophin and leukaemia inhibitory factor (LIF) in transplantation tolerance. Philos Trans R Soc Lond B Biol Sci 2005; 360:1687-94. [PMID: 16147533 PMCID: PMC1569543 DOI: 10.1098/rstb.2005.1697] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Immune self-tolerance is controlled by a subset of T lymphocytes that are regulatory (Treg) and epigenetically programmed to suppress auto-reactive immune effector cells in vivo. By extrapolation, donor-specific transplantation tolerance might be controlled by donor-specific Treg that have acquired the appropriate epigenetic program for tolerance. Although such tolerance has yet to be achieved in man, proof of concept comes from mouse models where regulatory transplantation tolerance can be induced within the complex micro-environment of the spleen or draining lymph node. By studying whole spleen cell populations in a murine model of transplantation tolerance we have incorporated a complexity of environmental factors when looking for specific features that characterize tolerance versus aggression. This approach has revealed unexpected patterns of gene activity in tolerance and most notably that a novel stem cell gene, axotrophin, regulates T lymphocyte responsiveness both in terms of proliferation and in release of leukaemia inhibitory factor (LIF). Since LIF is a regulator of stem cells in addition to being a key neuropoietic cytokine, these preliminary results linking both axotrophin and LIF to transplantation tolerance lead us to propose that regulatory pathways encoded during the epigenetic development of Treg cells are related to pathways that regulate fate determination of stem cells.
Collapse
|
48
|
Abstract
Ever since the discovery of Medawar, over 50 years ago, that immunological tolerance was an acquired phenomenon that could be manipulated in neonatal mice, the ability to induce therapeutic tolerance against autoantigens, allergens and organ grafts has been a major driving force in immunology. Within the last 20 years we have found that a brief treatment with monoclonal antibodies that block certain functional molecules on the surface of the T cell is able to reprogramme the established immune repertoire of the adult mouse, allowing indefinite acceptance of allografts or effective curing of autoimmune diseases. We are only now just beginning to define many of the regulatory mechanisms that induce and maintain the tolerant state with the aim of being able to safely and reliably apply these technologies to human clinical situations.
Collapse
|
49
|
Metcalfe SM, Muthukumarana PADS, Thompson HL, Haendel MA, Lyons GE. Leukaemia inhibitory factor (LIF) is functionally linked to axotrophin and both LIF and axotrophin are linked to regulatory immune tolerance. FEBS Lett 2005; 579:609-14. [PMID: 15670816 DOI: 10.1016/j.febslet.2004.12.027] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2004] [Revised: 11/29/2004] [Accepted: 12/08/2004] [Indexed: 11/27/2022]
Abstract
Axotrophin (axot) is a newly characterised stem cell gene and mice that lack axotrophin are viable and fertile, but show premature neural degeneration and defective development of the corpus callosum. By comparing axot+/+, axot+/- and axot-/- littermates, we now show that axotrophin is also involved in immune regulation. Both T cell proliferation and T cell-derived leukaemia inhibitory factor (LIF) were suppressed by axotrophin in a gene-dose-dependent manner. Moreover, a role for axotrophin in the feedback regulation of LIF is implicated. This is the first evidence that fate determination mediated by LIF maybe qualified by axotrophin.
Collapse
Affiliation(s)
- Su M Metcalfe
- Department of Surgery, University of Cambridge, Box 202, Level E9, Addenbrooke's Hospital, Hills Road, Cambridge CB2 2QQ, UK.
| | | | | | | | | |
Collapse
|
50
|
Bemelman F, Yong SL, Tissingh R, Schellekens P, ten Berge I. Suppression extends to major histocompatibility antigens linked to tolerizing minor histocompatibility antigens, but not the other way round. Transpl Int 2005; 18:423-8. [PMID: 15773962 DOI: 10.1111/j.1432-2277.2004.00056.x] [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: 11/26/2022]
Abstract
'Active suppression', a mechanism of transplantation tolerance, can spread to newly introduced minor antigens once these antigens are linked to tolerizing antigens. We explored whether this suppression can extend to major histocompatibility (MHC) antigens and whether this phenomenon can be demonstrated once tolerance is induced to a MHC antigen. Mice were tolerized using donor bone marrow plus CD4 and CD8 monoclonal antibodies. The following strain combinations were used: AKR (H-2k) into CBA (H-2k), a multiple minor difference and B6 (H-2b) into B6(bm12) (H-2b), a MHC class II difference. Tolerance was tested by a donorskingraft. CBA mice tolerant to AKR received a second skin carrying either AKR antigens plus additional multiple minor antigens [F1(AKRxBalb.K)] or carrying additional minors and a MHC class I antigen (B10.AKM-H2M). B6(bm12) (H-2b) tolerant to B6 (H-2b) were grafted with skin from a Balb.B donor (Balb minors linked to the tolerizing class II antigen) or from a B10.A(3R) strain (a MHC class I antigen linked to the tolerizing class II antigen). CBA mice tolerant to AKR accepted F1(AKRxBalb.K) skin, whereas F1(CBAxBalb.K) were rejected. Rejection of B10.AKM/H2M skin by tolerant mice was delayed as compared with nontolerant mice. Tolerant and nontolerant B6(bm12) mice rejected Balb.B skin and B10.A(3R) skin within the same time. Thus, in this model, suppression was linked to minors. Alloreactivity against minors and majors could be suppressed. Suppression linked to a class II antigen could not be demonstrated.
Collapse
|