1
|
Engelhart MJ, Glowacki RWP, Till JM, Harding CV, Martens EC, Ahern PP. The NQR Complex Regulates the Immunomodulatory Function of Bacteroides thetaiotaomicron. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:767-781. [PMID: 37486212 PMCID: PMC10527448 DOI: 10.4049/jimmunol.2200892] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 06/26/2023] [Indexed: 07/25/2023]
Abstract
The gut microbiome and intestinal immune system are engaged in a dynamic interplay that provides myriad benefits to host health. However, the microbiome can also elicit damaging inflammatory responses, and thus establishing harmonious immune-microbiome interactions is essential to maintain homeostasis. Gut microbes actively coordinate the induction of anti-inflammatory responses that establish these mutualistic interactions. Despite this, the microbial pathways that govern this dialogue remain poorly understood. We investigated the mechanisms through which the gut symbiont Bacteroides thetaiotaomicron exerts its immunomodulatory functions on murine- and human-derived cells. Our data reveal that B. thetaiotaomicron stimulates production of the cytokine IL-10 via secreted factors that are packaged into outer membrane vesicles, in a TLR2- and MyD88-dependent manner. Using a transposon mutagenesis-based screen, we identified a key role for the B. thetaiotaomicron-encoded NADH:ubiquinone oxidoreductase (NQR) complex, which regenerates NAD+ during respiration, in this process. Finally, we found that disruption of NQR reduces the capacity of B. thetaiotaomicron to induce IL-10 by impairing biogenesis of outer membrane vesicles. These data identify a microbial pathway with a previously unappreciated role in gut microbe-mediated immunomodulation that may be targeted to manipulate the capacity of the microbiome to shape host immunity.
Collapse
Affiliation(s)
- Morgan J. Engelhart
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Robert W. P. Glowacki
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jessica M. Till
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Clifford V. Harding
- Department of Pathology, Case Western Reserve University/University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Eric C. Martens
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Philip P. Ahern
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| |
Collapse
|
2
|
Danelli L, Cornish G, Merkenschlager J, Kassiotis G. Default polyfunctional T helper 1 response to ample signal 1 alone. Cell Mol Immunol 2021; 18:1809-1822. [PMID: 32313208 PMCID: PMC8245500 DOI: 10.1038/s41423-020-0415-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Accepted: 03/14/2020] [Indexed: 12/27/2022] Open
Abstract
CD4+ T cells integrate well-defined signals from the T-cell receptor (TCR) (signal 1) and a host of costimulatory molecules (signal 2) to initiate clonal expansion and differentiation into diverse functional T helper (Th) subsets. However, our ability to guide the expansion of context-appropriate Th subsets by deploying these signals in vaccination remains limited. Using cell-based vaccines, we selectively amplified signal 1 by exclusive presentation of an optimized peptide:MHC II (pMHC II) complex in the absence of classic costimulation. Contrary to expectations, amplified signal 1 alone was strongly immunogenic and selectively expanded high-affinity TCR clonotypes, despite delivering intense TCR signals. In contrast to natural infection or standard vaccines, amplified signal 1, presented by a variety of professional and nonprofessional antigen-presenting cells (APCs), induced exclusively polyfunctional Th1 effector and memory cells, which protected against retroviral infection and tumor challenge, and expanded tumor-reactive CD4+ T cells otherwise rendered unresponsive in tumor-bearing hosts. Together, our findings uncover a default Th1 response to ample signal 1 and offer a means to selectively prime such protective responses by vaccination.
Collapse
Affiliation(s)
- Luca Danelli
- Retroviral Immunology, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Georgina Cornish
- Retroviral Immunology, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
| | - Julia Merkenschlager
- Retroviral Immunology, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY, 10065, USA
| | - George Kassiotis
- Retroviral Immunology, The Francis Crick Institute, 1 Midland Road, London, NW1 1AT, UK.
- Department of Medicine, Faculty of Medicine, Imperial College London, London, W2 1PG, UK.
| |
Collapse
|
3
|
Donor chimera model for tolerance induction in transplantation. Hum Immunol 2013; 74:550-6. [PMID: 23354322 DOI: 10.1016/j.humimm.2013.01.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Revised: 12/09/2012] [Accepted: 01/14/2013] [Indexed: 12/14/2022]
Abstract
Tolerance induction is the basis of a successful transplantation with the goal being the re-establishment of homeostasis after transplantation. Non-autograft transplantation disrupts this maintenance drastically which would be avoided by administration of a novel procedure. At present, the blood group antigens and the genotypes of the donor and recipient are cross-matched before transplantation combined with a drug regimen that confers general immunosuppression. But the 'specific' unresponsiveness of the recipient to the donor organ, implied by 'tolerance', is not achieved in this process. This article introduces the 'donor chimera model' via the concept of the 'closed transplantation loop' approach for tolerance induction which seeks to limit the use of immunosuppressive therapy after transplantation.
Collapse
|
4
|
Andakyan A, Burruss S, Hong LS, Shen XD, Romanov S, Gao F, Feldman DM, Fishbein MC, Semiletova NV. Soluble donor-like MHC class I proteins induce CD4(+)CD25(+)CD8(-)FoxP3(+) cells with potential to ameliorate graft chronic injury. Transpl Immunol 2011; 24:203-9. [PMID: 21251981 DOI: 10.1016/j.trim.2011.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2010] [Revised: 12/03/2010] [Accepted: 01/08/2011] [Indexed: 11/29/2022]
Abstract
Conventional immunosuppressive therapies failed to prevent allograft chronic rejection. New approaches to modulate recipient immune response are needed. Donor-like MHC class I soluble proteins demonstrated therapeutic potential to suppress chronic rejection. The present study was designed to clarify the ability of MHC class I soluble proteins to induce T regulatory cells with true regulatory potential in a fully allogeneic rat cardiac transplant model. Donor-like MHC class I proteins upregulate small population of splenic CD8(-) negative CD4(+)CD25(+)FoxP3(+) positive cells. CD4(+) splenocytes after MHC therapy suppress lymphocyte proliferation against donor antigens in vitro. ACI recipients of WF hearts treated with CD4(+) cells, induced with donor-like MHC class I proteins (CD4-MHC), demonstrated stable survival of the transplanted organ (MST >120 days; n=17). Histology revealed that grafts of recipients treated with CD4-MHC had 23.6% vessels affected 100 days postgrafting. On the contrary, hearts obtained from long-term surviving hosts treated with CD4(+) cells induced with high-dose CsA (CD4-CsA) had 50-70% of affected vessels. CD4-MHC class I treated transplants were mostly CD3(-) negative, had low level of mast and FoxP3(+) cell infiltration compared to CD4-CsA treated hearts. Intragraft CD4(+) cells were close to mast cells in morphology. The same graft tissues had similar number of CD4(+) positive cells and mast cells suggesting existence of CD4(+) positive mast cells. On the other hand, a negligible number of FoxP3(+) positive cells in the grafts after CD4-MHC treatment supports the idea of CD4(+) positive FoxP3(+) negative mast cells population. We demonstrate that donor-like MHC class I protein therapy induces population of CD4(+)CD25(+)CD8(-)FoxP3(+) cells with potential to ameliorate development of transplant vascular disease and evoke CD4(+) positive FoxP3 negative mast cells in the secondary hosts.
Collapse
Affiliation(s)
- Arthur Andakyan
- UCLA School of Medicine, 10833 Le Conte Ave., 77-120 CHS, Los Angeles, CA 90095-7054, United States
| | | | | | | | | | | | | | | | | |
Collapse
|
5
|
|
6
|
Bishop KD, Harris JE, Mordes JP, Greiner DL, Rossini AA, Czech MP, Phillips NE. Depletion of the programmed death-1 receptor completely reverses established clonal anergy in CD4(+) T lymphocytes via an interleukin-2-dependent mechanism. Cell Immunol 2009; 256:86-91. [PMID: 19230866 DOI: 10.1016/j.cellimm.2009.01.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2008] [Revised: 01/16/2009] [Accepted: 01/22/2009] [Indexed: 12/21/2022]
Abstract
Recent studies have implicated the cell surface receptor Programmed Death-1 (PD-1) in numerous models of T cell anergy, though the specific mechanisms by which the PD-1 signal maintains tolerance is not clear. We demonstrate that the depletion of PD-1 with siRNA results in a complete reversal of clonal anergy in the A.E7 T cell model, suggesting that the mechanism by which PD-1 maintains the anergic phenotype is a T-cell-intrinsic phenomenon, and not one dependent on other cell populations in vivo. We have also shown that the neutralization of IL-2 during restimulation abrogates the effect of PD-1 depletion, suggesting that tolerance mediated by PD-1 is wholly IL-2 dependent, and likewise intrinsic to the tolerized cells.
Collapse
Affiliation(s)
- Kenneth D Bishop
- University of Massachusetts Medical School, Worcester, 01655, USA
| | | | | | | | | | | | | |
Collapse
|
7
|
Doraiswamy A, Narayan RJ, Harris ML, Qadri SB, Modi R, Chrisey DB. Laser microfabrication of hydroxyapatite-osteoblast-like cell composites. J Biomed Mater Res A 2007; 80:635-43. [PMID: 17051538 DOI: 10.1002/jbm.a.30969] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
We have developed a novel approach for layer-by-layer growth of tissue-engineered materials using a direct writing process known as matrix assisted pulsed laser evaporation direct write (MAPLE DW). Unlike conventional cell-seeding methods, this technique provides the possibility for cell-material integration prior to artificial tissue fabrication. This process also provides greater flexibility in selection and processing of scaffold materials. In addition, MAPLE DW offers rapid computer-controlled deposition of mesoscopic voxels at high spatial resolutions. We have examined MAPLE DW processing of zirconia and hydroxyapatite scaffold materials that can provide a medical device with nearly inert and bioactive implant-tissue interfaces, respectively. We have also demonstrated codeposition of hydroxyapatite, MG 63 osteoblast-like cells, and extracellular matrix using MAPLE DW. We have shown that osteoblast-like cells remain viable and retain the capacity for proliferation when codeposited with bioceramic scaffold materials. Our results on MG 63-hydroxyapatite composites can be extended to develop other integrated cell-scaffold structures for medical and dental applications.
Collapse
Affiliation(s)
- A Doraiswamy
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | | | | | | | | | | |
Collapse
|
8
|
Karaji AG. Application of popliteal lymph node assay to evaluate tolerogenic effect of donor leukocyte infusion. Transpl Immunol 2006; 16:20-4. [PMID: 16701172 DOI: 10.1016/j.trim.2006.03.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2006] [Accepted: 03/09/2006] [Indexed: 11/17/2022]
Abstract
BACKGROUND Popliteal lymph node assay (PLNA) has long been proposed to detect immunostimulating potential of chemicals. Here, the PLNA was used to evaluate the effect of donor leukocyte infusion on recipients' reaction to donor-specific antigens. METHODS Donor rats' peripheral blood leukocytes (ranging from 1 x 10(4) to 500 x 10(4) cells) were intravenously (i.v.) infused into recipients. A week later recipients' reaction to donor-specific antigen was evaluated, using the PLNA technique, by subcutaneous injection of donor spleen cells to one hind footpad of recipients and injection of saline to the other. Seven days later all recipients were killed and their PLNs' weight and cellularity indices were determined. While the same process was applied to the positive control (PC) animals, rats without leukocyte infusion, negative control (NC) animals, rats without leukocyte infusion, were injected in both hind footpads with saline. RESULTS The PLN weight indices of recipients of: > or =5 x 10(4) leukocytes were significantly lower than PC animals (P < 0.001), whereas the weight indices of recipients of 1 x 10(4) cells were similar to PC group but higher than NC animals (P < 0.0001). However, the PLN cellularity indices of recipients of < or =10 x 10(4) cells were not different from PC animals but the PLN cellularity indices of recipients of: > or =50 x 10(4) cells were significantly lower than PC group (P < 0.05). CONCLUSION Overall, these results suggest that donor leukocytes infusion dose-dependently decrease reaction to donor-specific antigens, but a state of tolerance to donor antigen might be induced at the dose of: > or =50 x 10(4) cells. PLNA appears to represent a simple test model to quantify efficacy of immunotolerance protocols.
Collapse
Affiliation(s)
- Ali Gorgin Karaji
- Department of Microbiology and Immunology, School of Medicine, Kermanshah University of Medical Sciences, Iran.
| |
Collapse
|
9
|
Lowsky R, Negrin RS. Total lymphoid irradiation and transplantation tolerance. Curr Opin Organ Transplant 2006. [DOI: 10.1097/01.mot.0000203882.10310.65] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
10
|
Tian C, Bagley J, Forman D, Iacomini J. Induction of central tolerance by mature T cells. THE JOURNAL OF IMMUNOLOGY 2005; 173:7217-22. [PMID: 15585843 DOI: 10.4049/jimmunol.173.12.7217] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Induction of immunological tolerance is highly desirable for the treatment and prevention of autoimmunity, allergy, and organ transplant rejection. Adoptive transfer of MHC class I disparate mature T cells at the time of reconstitution of mice with syngeneic bone marrow resulted in specific tolerance to allogeneic skin grafts that were matched to the T cell donor strain. Mature allogeneic T cells survived long-term in reconstituted hosts and were able to re-enter the thymus. Analysis of T cell development using transgenic mice expressing an alloantigen-reactive TCR revealed that expression of allogeneic MHC class I on adoptively transferred mature T cells mediated negative selection of developing alloreactive T cells in the thymus. Thus, mature allogeneic T cells are able to mediate central deletion of alloreactive cells and induce transplantation tolerance without the requirement for any other alloantigen-expressing cell type.
Collapse
Affiliation(s)
- Chaorui Tian
- Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129
| | | | | | | |
Collapse
|
11
|
Abstract
Clinical transplantation tolerance has remained an elusive goal in the 50 yr since it was first described in experimental animals. Greater understanding of the molecular mechanisms responsible for allorecognition have allowed for the development of promising immunosuppressive strategies that may bring us closer to reproducible induction of tolerance; consideration of past successes and failures from both clinical and basic science is required to define future challenges facing this field. This article reviews mechanisms of self and transplantation tolerance, translation of basic science research to clinical protocols in animals and human beings, the changing role of immunosuppression, complications following tolerance induction and controversies surrounding the choice of patients for tolerance trials with a focus on issues relevant to pediatric patients. The role of the Immune Tolerance Network is discussed along with realistic goals for tolerance induction in human beings over the next decade.
Collapse
Affiliation(s)
- Kathryn J Tinckam
- Transplantation Research Center, Brigham and Women's Hospital and Children's Hospital Boston, Harvard Medical School, Boston, MA 02115, USA
| | | |
Collapse
|
12
|
Zhao RC, Liao L, Han Q. Mechanisms of and perspectives on the mesenchymal stem cell in immunotherapy. ACTA ACUST UNITED AC 2004; 143:284-91. [PMID: 15122172 DOI: 10.1016/j.lab.2003.11.009] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Mesenchymal stem cells (MSCs) are an important cell population in the bone-marrow microenvironment and are considered to be engaged mainly in the support of hematopoiesis. Recent work has shown that MSCs also have profound immunomodulatory function, both in vitro and in vivo. Because MSCs can be expanded rapidly to the numbers required for clinical application, several preclinical and clinical studies have been performed in the areas of immune diseases and bone-marrow transplantation. In this review we discuss the mechanisms underlying the MSC's immunomodulating properties and its potential applications.
Collapse
Affiliation(s)
- Robert Chunhua Zhao
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, People's Republic of China.
| | | | | |
Collapse
|
13
|
Tahara H, Iwanami N, Tabata N, Matsumura H, Matsuura T, Kurita T, Miyazawa M. Both T and non-T cells with proliferating potentials are effective in inducing suppression of allograft responses by alloantigen-specific intravenous presensitization combined with suboptimal doses of 15-deoxyspergualin. Transpl Immunol 2004; 13:25-32. [PMID: 15203125 DOI: 10.1016/j.trim.2004.01.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2003] [Accepted: 01/21/2004] [Indexed: 11/15/2022]
Abstract
In an MHC class I-disparate combination of mouse strains, a single intravenous injection of donor spleen cells combined with 10 suboptimal doses of 15-deoxyspergualin (DSG) administration was effective in inducing donor-specific suppression of cytotoxic T-lymphocyte (CTL) responses and prolonged survival of the relevant skin allograft. Proliferative potentials of the donor spleen cells were requirement for the induction of suppressed allospecific responses, but both highly purified T cells and non-T cells were equally effective to induce the suppression of CTL responses by intravenous injection. These results have shown that, although working on different mechanisms, DSG is as effective as FK506 or rapamycin in inducing allograft tolerance when used at suboptimal doses along with the donor-specific intravenous presensitization, and an immune mechanism other than well-characterized veto T cells is working in this model in suppressing alloreactive CTL precursors.
Collapse
Affiliation(s)
- Hideo Tahara
- Department of Immunology, Kinki University School of Medicine, Osaka-Sayama, Osaka 589-8511, Japan
| | | | | | | | | | | | | |
Collapse
|
14
|
Fujisawa K, Saito S, Okada Y, Fujiwara T, Yagi T, Iwagaki H, Tanaka N. Suppression of allogeneic response by viral IL-10 gene transfer. Cell Transplant 2004; 12:379-87. [PMID: 12911125 DOI: 10.3727/000000003108746920] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Th1 cell activation and cytokine production shift the balance between Th1 and Th2, favoring the upregulation of proinflammatory activity that leads to destruction of allogeneic hepatocytes following transplantation. Th2-type cytokines. such as IL-10, have immune regulatory function. The aim of this study was to determine the antirejection efficacy of allogeneic hepatocytes with spheroidal shape (spheroids) genetically modified with viral IL-10 (vIL-10). Allogeneic hepatocyte spheroids, transferred vIL-10 gene by using adenovirus as the vector, were transplanted into the spleen of Nagase's analbuminemic rats (NAR). NAR transplanted with vIL-10-transfected hepatocytes showed an abrupt rise in serum albumin levels that peaked on day 7 and remained at high levels up to day 21 after transplantation. The peak level of albumin on day 7 in vIL-10-transfected NAR was eminently higher than that in nontransfected NAR. Histopathological analysis revealed that in nontransfected NAR hepatocyte spheroids were more or less rejected on day 4, and, in contrast, vIL-10-transfected spheroids were still not rejected on day 14. This protective effect correlated with sustained high vIL-10 level in the splenic vein in NAR transplanted with vIL-10-transfected hepatocyte spheroids, suggesting that vIL-10 secreted from the transplanted hepatocytes induced an active suppression of allogeneic response. This study provides evidence to support the possibility of using vIL-10 gene therapy to prevent allogeneic response in hepatocyte transplantation.
Collapse
Affiliation(s)
- Kenji Fujisawa
- Department of Gastroenterological Surgery and Transplant, Okayama University Graduate School of Medicine and Dentistry, Okayama city, 700-8558 Japan
| | | | | | | | | | | | | |
Collapse
|
15
|
Muramatsu K, Valenzuela RG, Bishop AT. Detection of chimerism following vascularized bone allotransplantation by polymerase chain reaction using a Y-chromosome specific primer. J Orthop Res 2003; 21:1056-62. [PMID: 14554219 DOI: 10.1016/s0736-0266(03)00108-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Chimerism following allogeneic organ transplantation is a phenomenon known to occur and be associated with development of immunologic tolerance in allotransplantation. However, little is known about graft cell migration following vascularized bone allografting. In this study, chimerism was assessed following vascularized tibia transplantation from male DA or PVG donors to female PVG rat recipients using a semi-quantitative polymerase chain reaction for the Y-chromosome. FK-506 (Tacrolimus) was administered after transplantation for immunosuppression. All immunosuppresssed PVG rat recipients of PVG bone grafts showed a high level of chimerism (1%) in the thymus, spleen, liver and cervical lymph nodes at 18 weeks post-transplant. Donor cells were also detected in the contralateral tibia and humerus. In non-immunosuppressed PVG rat recipients of DA bone grafts, donor cells were detected in the spleen in three of five rats within 2 weeks post-transplant. In these animals the bone grafts were severely rejected. In immunosuppressed PVG rat recipients of DA bone grafts, two of five, four of eight and eight of 10 rats showed low level chimerism (0.1%) in peripheral blood at 1, 12, and 18 weeks post-transplant. Six rats showed a high level of chimerism in the spleen and thymus. Histological studies revealed no rejection findings through 18 weeks post-transplant. Our results indicate that chimerism, or the presence of graft cells in host tissue, may occur in the face of acute rejection and be demonstrable following vascularized isograft and allograft living bone transplantation when chronic immunosuppression is maintained. Graft vascular patency during the short-term likely allows cellular migration, even in the face of acute rejection. Long-term survival and proliferation of graft marrow elements in host tissue may be possible with adequate immunosuppression.
Collapse
Affiliation(s)
- Keiichi Muramatsu
- Department of Orthopaedic Surgery, Mayo Clinic, 200 First Street, S.W., Rochester, MN 55905, USA
| | | | | |
Collapse
|
16
|
Kim DW, Chung YJ, Kim TG, Kim YL, Oh IH. Cotransplantation of third-party mesenchymal stromal cells can alleviate single-donor predominance and increase engraftment from double cord transplantation. Blood 2003; 103:1941-8. [PMID: 14592834 DOI: 10.1182/blood-2003-05-1601] [Citation(s) in RCA: 121] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Although the infusion of umbilical cord blood (UCB) from multiple donors can be a strategy to overcome the cell dose limitation frequently encountered in UCB transplantation, clinical trials have revealed that cells from one donor dominate engraftment. To investigate the origin of and the factors influencing this inequality, we performed mixed transplantation of 2 UCB units with varying degrees of HLA disparities into NOD/SCID mice and determined donor origins by polymerase chain reaction-sequence-specific oligonucleotide probe (PCR-SSOP) or real-time quantitative (RQ)-PCR for human short tandem repeats (STRs). When total mononuclear cells from 2 units were transplanted as a mixture, cells from one donor predominated (ratio, 81:19), despite comparable overall engraftment when infused as single units, and no augmentation in overall engraftment was observed when compared with the single-unit controls. However, lineage depletion or cotransplantation of mesenchymal stromal cells (MSCs) expanded from third-party bone marrow resulted in more balanced coengraftment. Direct comparison of double UCB transplantation in the presence or absence of MSCs showed that the reduced deviation in the donor ratio (1.8:1 vs. 2.8:1) correlated with a higher overall level of engraftment with MSC cotransplantation. These results indicate that third-party MSCs can be used to alleviate donor deviation and to facilitate engraftment of multidonor UCB.
Collapse
Affiliation(s)
- Dong-Wook Kim
- Cell and Gene Therapy Institute and the Department of Cellular Medicine and Biology, The Catholic University of Korea, Seoul, Korea
| | | | | | | | | |
Collapse
|
17
|
Affiliation(s)
- Fadi G Lakkis
- Section of Nephrology, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06520-8029, USA.
| | | |
Collapse
|
18
|
Kopp T, Lenz P, Bello-Fernandez C, Kastelein RA, Kupper TS, Stingl G. IL-23 production by cosecretion of endogenous p19 and transgenic p40 in keratin 14/p40 transgenic mice: evidence for enhanced cutaneous immunity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:5438-44. [PMID: 12759419 DOI: 10.4049/jimmunol.170.11.5438] [Citation(s) in RCA: 103] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
p40, the common subunit of the proinflammatory cytokines IL-12 and IL-23, is produced by resident skin cells. Whereas the in vivo effects of IL-12 are well established, little is known about the role of IL-23 in cutaneous immune responses. In this study we show that p40 transgenic (TG) mice constitutively produce IL-23 (p19/p40), but not IL-12 (p35/p40), in basal keratinocytes by cosecretion of TG p40 with endogenous p19. Repeated injections of rIL-23 in littermate (LM) mice result in an inflammatory skin disease similar to that of p40 TG mice, confirming the proinflammatory activity of IL-23. Furthermore, IL-23 secretion by p40 TG keratinocytes induces elevated numbers of Langerhans cells (LC) with a marked up-regulation of costimulatory molecules, indicating advanced maturation of keratin 14 (K14)/p40 LC when compared with LM LC. At the functional level, freshly isolated K14/p40 LC greatly exceeded LC from LM animals in their capacity to stimulate allogeneic T cell proliferation. To assess whether IL-23 regulates cutaneous immune responses in vivo, we used an allogeneic skin transplantation model. Full thickness skin grafts from K14/p40 donors (H-2(q)) transplanted across a MHC class I and class II barrier onto BALB/c (H-2(d)) recipients were rejected in a significantly accelerated fashion (mean survival time: 8.8 days) when compared with skin grafts from non-TG LM (H-2(q)) (mean survival time: 10.7 days, p < 0.01). Based on these results we propose that IL-23-induced changes of LC may be an important mechanism in directing the outcome of cutaneous immune responses.
Collapse
Affiliation(s)
- Tamara Kopp
- Department of Dermatology, Division of Immunology, Allergy and Infectious Diseases, University of Vienna Medical School, Vienna International Research Cooperation Center, Vienna, Austria.
| | | | | | | | | | | |
Collapse
|
19
|
Abstract
Advances in transplantation biology have greatly improved patient outcomes following transplant surgery. However, generalized immunosuppression remains the Achilles heel of modern transplantation surgery with its associated infectious and neoplastic morbidities. Tolerance remains the ultimate goal for the entire field. Although recent advances in transplant immunology suggest that tolerance may be achievable in the near future, the complex and redundant nature of the human immune system may not allow us to circumvent such a basic function as the recognition of nonself. In this paper, advances in transplant immunology are reviewed and their potential relevance to achieving the "Holy Grail" of transplantation are discussed.
Collapse
Affiliation(s)
- Rebecca A Schroeder
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | | | | |
Collapse
|
20
|
Tian C, Bagley J, Iacomini J. Expression of antigen on mature lymphocytes is required to induce T cell tolerance by gene therapy. THE JOURNAL OF IMMUNOLOGY 2002; 169:3771-6. [PMID: 12244171 DOI: 10.4049/jimmunol.169.7.3771] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Expression of a retrovirally encoded allogeneic MHC class I gene in bone marrow-derived cells can be used to induce tolerance to the product of the retrovirally transduced gene. In this work we examined whether expression of a retrovirally transduced allogeneic MHC class I gene in bone marrow-derived cells from recombinase-activating gene-1 (RAG-1)-deficient mice was sufficient to induce tolerance when transplanted into conditioned hosts together with bone marrow from MHC-matched wild-type mice. Reconstitution of mice with either MHC-matched RAG-1-deficient or wild-type bone marrow transduced with the allogeneic MHC class I gene H-2K(b) led to long-term expression of K(b) on the surface of bone marrow-derived hematopoietic lineages. T cells from mice reconstituted with H-2K(b)-transduced wild-type bone marrow were tolerant to K(b). In contrast, expression of K(b) in the periphery of mice reconstituted with a mixture of retrovirally transduced RAG-1-deficient bone marrow and mock-transduced wild-type bone marrow fell below detectable levels by 4 wk after transplantation. T cells that developed in these mice appeared to be hyporesponsive to K(b), demonstrating that expression of K(b) on bone marrow-derived APCs was not sufficient to induce tolerance. Our data suggest that induction of tolerance in molecular chimeras requires expression of the retrovirally transduced allogeneic MHC Ag on the surface of mature lymphocytes that populate the host thymus.
Collapse
Affiliation(s)
- Chaorui Tian
- Transplantation Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA
| | | | | |
Collapse
|
21
|
Affiliation(s)
- K Muramatsu
- Department of Orthopedic Surgery and Microvascular Research, Mayo Clinic, Rochester, Minnesota 55905, USA
| | | |
Collapse
|
22
|
Ciancio G, Burke GW, Garcia-Morales R, Suzart K, Rosen A, Ricordi C, Kenyon NS, Mathew JM, Tzakis AG, Esquenazi V, Miller J. Effect of living-related donor bone marrow infusion on chimerism and in vitro immunoregulatory activity in kidney transplant recipients. Transplantation 2002; 74:488-96. [PMID: 12352907 DOI: 10.1097/00007890-200208270-00010] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND In a previously reported series of donor-specific bone marrow cell (DBMC) infusions in cadaver kidney transplant recipients, there appeared to be an improvement in long-term graft survival (6 years) and fewer chronic rejections, which correlated with increasing DBMC chimerism (approximately 1.4% in the iliac crest bone marrow compartment now at 6 years). Prompted by this, we embarked on a study of DBMC infusion in living-related donor (LRD) kidney transplant recipients. METHODS Between November 1996 and May 2000, 47 LRD kidney transplant recipients received donor iliac crest marrow (1.8 x 10(8)+/-1.9 x 10(8) cells/kg body weight+/-SD) in a single infusion 4 days postoperatively. Either OKT3 (n=26) or daclizumab (n=21) were used for induction therapy, with maintenance tacrolimus, mycophenolate mofetil, and methylprednisolone immunosuppression. These recipients were prospectively compared with 39 noninfused LRD kidney transplants (control group), which received equivalent immunosuppression in the same time period. Clinical follow-up ranged from 19.0 months to 61.6 months (mean 33.2 months). Polymerase chain reaction-flow chimerism analysis and in vitro assays of immunoregulatory activity of chimeric cells were performed. RESULTS The incidence of acute rejection over this period of time was 10.6% and 10.3%, respectively (i.e., did not differ between groups). Immunosuppressive dosages were somewhat (but not statistically) lower over time in the DBMC group. Four-year actuarial patient and graft survival for the DBMC-infused group was 98% and 98%, and 98% and 95% for the control group, respectively ( =NS). DBMC infusion was well tolerated, with no increase in infectious episodes. DBMC chimerism in recipient iliac crest marrow has increased more rapidly than might be predicted from results previously seen in the cadaver group, despite four times fewer DBMC infused. DBMCs and (donor) peripheral blood mononuclear cells purified by immunobeads from recipient blood or bone marrow (recipient-derived donor cells) inhibited mixed leukocyte responses of the recipient to the donor more strongly than freshly obtained peripheral blood cells drawn from the donors or even compared with bone marrow cells aspirated from the donors in a previously reported group of experiments. Additionally, similarly purified recipient-derived recipient cells from the same chimeric recipient more strongly inhibited the same mixed leukocyte response reactions autologously than a large group of nonchimeric (autologous) bone marrow modulating cells in similar reactions. CONCLUSIONS These observations confirm that an immunoregulatory process appears to have been generated by DBMC infusion, encouraging a further decrease in immunosuppressive dosing using such assays in the future.
Collapse
Affiliation(s)
- Gaetano Ciancio
- Department of Surgery, Division of Transplantation, University of Miami School of Medicine, Miami, FL 33101, USA. E-mail:
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Yang H, Thomas D, Boffa DJ, Ding R, Li B, Muthukumar T, Sharma VK, Lagman M, Luo GX, Kapur S, Liou HC, Hancock WW, Suthanthiran M. Enforced c-REL deficiency prolongs survival of islet allografts1. Transplantation 2002; 74:291-8. [PMID: 12177605 DOI: 10.1097/00007890-200208150-00002] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND The NF-kappaB/Rel family of transcription factors regulates biologic processes ranging from apoptosis to inflammation and innate immunity. Whether c-Rel, a lymphoid-predominant member of the NF-kappaB/Rel family, is essential for transplantation immunity is not known. METHODS We explored the role of c-Rel in the anti-allograft repertory using mice with targeted disruption of the c-Rel gene (c-Rel-/-) as recipients of H-2 mismatched islet allografts. Allogeneic DBA/2 (H-2d) islets were transplanted into the renal subcapsular space of diabetic c-Rel-/- C57BL/6 (H-2b) mice or the c-Rel +/+ C57BL/6 wild-type mice. Islet graft survival, cellular traffic into the islet grafts and their phenotype, and intragraft expression of cytokines and cytotoxic attack molecules were determined at the protein (by immunohistochemistry) and mRNA (by real-time quantitative polymerase chain reaction) levels. RESULTS We found superior islet graft survival in the c-Rel-/- recipients compared to c-Rel+/+ C57BL/6 recipients. Splenocytes from c-Rel-/- mice proliferated poorly compared to splenocytes from the c-Rel+/+ mice on stimulation with anti-CD3 mAbs or Con A. Peri-islet infiltration composed of T lymphocytes and macrophages was found in both c-Rel+/+ recipients and c-Rel-/- recipients, but intra-islet infiltration was observed only in c-Rel+/+ recipients. Immunohistologic and molecular studies showed impaired T helper-type 1 immunity and decreased intragraft expression of cytotoxic attack molecules perforin and granzyme B in c-Rel-/- recipients as compared to wild-type recipients. CONCLUSIONS Our results demonstrate that c-Rel is essential for robust rejection of islet allografts and support the idea that strategies that impair c-Rel function may be of value for constraining alloimmunity and facilitating survival of allogafts.
Collapse
Affiliation(s)
- Hua Yang
- Division of Nephrology, Department of Medicine, Weill Medical College of Cornell University, New York, NY, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Affiliation(s)
- D H Sachs
- Transplantation Biology Research Center, Massachusetts General Hospital/Harvard Medical School, Department of Medicine, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, Massachusetts 02129, USA
| | | | | | | |
Collapse
|
25
|
Frasca L, Scottà C, Lombardi G, Piccolella E. Human anergic CD4+ T cells can act as suppressor cells by affecting autologous dendritic cell conditioning and survival. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:1060-8. [PMID: 11801639 DOI: 10.4049/jimmunol.168.3.1060] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
T cell suppression exerted by regulatory T cells represents a well-established phenomenon, but the mechanisms involved are still a matter of debate. Recent data suggest that anergic T cells can suppress responder T cell activation by inhibiting Ag presentation by dendritic cells (DC). In this study, we focused our attention on the mechanisms that regulate the susceptibility of DC to suppressive signals and analyzed the fate of DC and responder T cells. To address this issue, we have cocultured human alloreactive or Ag-specific CD4+ T cell clones, rendered anergic by incubation with immobilized anti-CD3 Ab, with autologous DC and responder T cells. We show that anergic T cells affect either Ag-presenting functions or survival of DC, depending whether immature or mature DC are used as APC. Indeed, MHC and costimulatory molecule expression on immature DC activated by responder T cells is inhibited, while apoptotic programs are induced in mature DC and in turn in responder T cells. Ligation of CD95 by CD95L expressed on anergic T cells in the absence of CD40-CD40L (CD154) interaction are critical parameters in eliciting apoptosis in both DC and responder T cells. In conclusion, these findings indicate that the defective activation of CD40 on DC by CD95L+ CD154-defective anergic T cells could be the primary event in determining T cell suppression and support the role of CD40 signaling in regulating both conditioning and survival of DC.
Collapse
Affiliation(s)
- Loredana Frasca
- Department of Cellular and Developmental Biology, La Sapienza University, Via dei Sardi, 70-00185 Rome, Italy
| | | | | | | |
Collapse
|
26
|
Abstract
BACKGROUND Recipient type mononuclear cells infiltrating kidney allografts have different phenotypes and functions according to the fate of the graft. We hypothesized that different genetic programs were involved in rejected or accepted tissues and thus, transcripts that correlated with the clinical status could be identified by a differential expression strategy. This strategy was applied to miniature swine class II matched, class I disparate kidney grafts, which are accepted in recipient animals treated for 12 days with Cyclosporin A (CsA). METHODS The mRNA differential display RT-PCR technique (DDRT-PCR) was used to detect clinical status-specific transcripts. cDNA templates for this analysis were derived from biopsies of accepted (CsA treated) and rejected (untreated) kidney grafts 8 days post-transplantation. RESULTS A first screening procedure identified 23 PCR products differentially amplified in either tolerant or rejector samples. Nucleotide sequence of these partial transcripts showed that 11 out of 23 (48%) sequences had unknown open reading frames while 12 had substantial homology to known sequences. To validate the approach, rejection-associated (RA) cDNA 1 (RA-1) was characterized further. The results indicated that RA-1 is the porcine equivalent of secreted protein acidic and rich in cysteine (SPARC). Expression studies demonstrated that upregulation of SPARC gene transcription preceded other indicators of kidney dysfunction and correlated with the extent of graft infiltration. CONCLUSION DDRT-PCR appears to be a powerful technique to identify genes differentially expressed in grafted tissues that correlate with tolerance or rejection. One of the gene transcripts identified through this method, SPARC, may be a reliable marker of tissue injury consequent to cellular infiltration and rejection.
Collapse
Affiliation(s)
- Isabel M McMorrow
- Transplantation Biology Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, Massachusetts 02129, USA
| | | | | | | |
Collapse
|
27
|
Abstract
BACKGROUND Copolymer 1 (Cop 1) was previously shown to prevent graft-versus-host disease and interfere in various manifestations of immune rejection. In this study, we tested whether Cop 1 can also hinder the reactivity of host against graft and inhibit graft rejection. METHODS Cop 1 was tested in two transplantation systems: skin and thyroid grafting assays. The effect of Cop 1 on T cell reactivity was investigated by proliferation and cytokine secretion of spleen and lymph node cells from transplanted mice, as well as the T cell lines generated therefrom. RESULTS Cop 1 treatment prolonged skin graft survival and inhibited the functional deterioration of thyroid grafts, in various strain combinations, across minor and major histocompatibility barriers, similarly to the potent immunosuppressive drug FK506. Cop 1 inhibited the proliferation of graft-specific T cell lines, as well as their interleukin (IL)-2 and interferon-gamma (IFN-gamma) secretion, when incubated in vitro with the stimulating allogeneic cells. Spleen and lymph node cells from Cop 1-treated mice, as well as the T cell lines generated from them, demonstrated a pronounced inhibition of proliferation and secretion of T helper (Th)1 cytokines in response to graft cells. In addition, cells from Cop 1-treated mice secreted high amounts of Th2 cytokines in response to Cop 1 and small but significant amounts of Th2 cytokines, mainly IL-10, in response to allograft cells. CONCLUSIONS Cop 1 treatment inhibited the Th1 response to graft and induced a Th2 cytokines secretion in response to both Cop 1 and graft cells, leading to improved survival and function of the transplanted grafts.
Collapse
Affiliation(s)
- R Aharoni
- Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel
| | | | | | | |
Collapse
|
28
|
Zhai Y, Li J, Hammer M, Busuttil RW, Volk HD, Kupiec-Weglinski JW. Evidence of T cell clonality in the infectious tolerance pathway: implications toward identification of regulatory T cells. Transplantation 2001; 71:1701-8. [PMID: 11455246 DOI: 10.1097/00007890-200106270-00001] [Citation(s) in RCA: 20] [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
We have shown that a rare population of regulatory CD4+ T cells plays a key role in the acquisition of infectious tolerance in rat sensitized recipients of cardiac allografts pretreated with nondepleting anti-CD4 mAb. This study was designed to analyze the TCR Vbeta expression patterns in this transplantation model. First, we used Vbeta-specific RT-PCR to show that there was no differential usage of TCR Vbeta genes by T cells mediating rejection or tolerance. Indeed, graft-infiltrating lymphocytes expressed most of the 22 known rat TCR Vbeta genes in both recipient groups, suggesting unrestricted TCR Vbeta repertoire in alloreactive T cells. Then, we applied CDR3 spectrotyping of TCR beta-chain to assess the clonality of T cells at different anatomic sites. CDR3 size restriction, indicative of the presence of T cell clones, was observed in graft-infiltrating lymphocytes but not in draining lymph nodes or spleen of tolerant hosts. Consisent with the clonal expansion, T cells in tolerated grafts exhibited the memory phenotype at a much higher percentage as compared with peripheral lymphoid organs. Moreover, in tolerated graft-infiltrating lymphocytes, the CD3 size restriction occurred in limited Vbeta gene families, with Vbeta8.1 and Vbeta18 most frequently detected. Hence, T cells at the graft site of tolerant recipients contain T cell clones expressing selective Vbeta genes. This phenotypic characteristics of the tolerogenic GILs may potentially be used as a novel marker to identify operational regulatory T cells in organ allograft recipients.
Collapse
Affiliation(s)
- Y Zhai
- The Dumont-UCLA Transplant Center, Rm. 77-120 CHS, 10833 Le Conte Ave, Los Angeles, CA 90095, USA
| | | | | | | | | | | |
Collapse
|
29
|
Horuzsko A, Lenfant F, Munn DH, Mellor AL. Maturation of antigen-presenting cells is compromised in HLA-G transgenic mice. Int Immunol 2001; 13:385-94. [PMID: 11222508 DOI: 10.1093/intimm/13.3.385] [Citation(s) in RCA: 90] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The human MHC class Ib antigen HLA-G is thought to regulate maternal immune responses during pregnancy. Here we show that expression of HLA-G in transgenic mice diminished cellular immunity by inhibiting maturation of myelomonocytic cells into functional antigen-presenting cells (APC). Skin allografts applied to HLA-G transgenic mice survived longer and resultant T cell responses were less potent compared to control mice. T cells from HLA-G mice responded normally to allogeneic APC and immunohistological analyses of spleen revealed no marked abnormalities. However, spontaneous outgrowths of myeloid cells were observed when bone marrow or splenocytes from HLA-G mice were cultured in vitro, but functionally competent APC did not develop spontaneously in bone marrow cultures supplemented with granulocyte macrophage colony stimulating factor (GM-CSF). Addition of lipopolysaccharide (LPS) to GM-CSF-derived bone marrow cultures rescued APC maturation. Studies using HLA-G tetrameric reagents revealed that HLA-G-specific binding activity was associated with CD11c(+) myelomonocytic cells, while binding to lymphoid and NK cell subsets was undetectable. These data show that spontaneous maturation of functionally competent dendritic cells (DC) is compromised in HLA-G mice. We hypothesize that HLA-G inhibits maturation of DC via receptor-mediated interactions with myelomonocytic precursors, which render immature DC precursors unable to receive signals from activated T cells.
Collapse
MESH Headings
- Animals
- Antigen-Presenting Cells/pathology
- Biopolymers
- Bone Marrow Cells/pathology
- Cell Differentiation/drug effects
- Cells, Cultured/drug effects
- Cytotoxicity, Immunologic
- Dendritic Cells/pathology
- Graft Survival
- Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology
- HLA Antigens/chemistry
- HLA Antigens/genetics
- HLA Antigens/physiology
- HLA-G Antigens
- Histocompatibility Antigens Class I/chemistry
- Histocompatibility Antigens Class I/genetics
- Histocompatibility Antigens Class I/physiology
- Histones/chemistry
- Immunity, Cellular
- Immunologic Deficiency Syndromes/genetics
- Immunologic Deficiency Syndromes/immunology
- Immunologic Deficiency Syndromes/pathology
- Killer Cells, Natural/immunology
- Lymphocyte Activation
- Lymphocyte Culture Test, Mixed
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C3H
- Mice, Inbred C57BL
- Mice, Inbred CBA
- Mice, Transgenic
- Myeloid Cells/drug effects
- Myeloid Cells/pathology
- Peptide Fragments/metabolism
- Protein Folding
- Recombinant Fusion Proteins/physiology
- Skin Transplantation/immunology
- Spleen/pathology
- Transplantation, Homologous/immunology
- beta 2-Microglobulin/chemistry
Collapse
Affiliation(s)
- A Horuzsko
- Program in Molecular Immunology, Department of Medicine, Medical College of Georgia, Augusta, GA 30912, USA
| | | | | | | |
Collapse
|
30
|
Nakagawa K, Matsuno T, Iwagaki H, Morimoto Y, Fujiwara T, Sadamori H, Inagaki M, Urushihara N, Yagi T, Tanaka N. Donor dendritic cells and recipient Kupffer cells in the induction of donor-specific immune hyporesponsiveness. J Int Med Res 2001; 29:119-30. [PMID: 11393345 DOI: 10.1177/147323000102900209] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The aim of this study was to investigate the ability of portovenously administered donor antigens to induce immune hyporesponsiveness. Lewis (LEW, RT-1l) rats received Brown Norway (BN, RT-1n) rat donor splenocytes, via either the portal vein (PV group) or the peripheral vein (IV group). The immune responses of LEW rats, treated with either donor BN or third party Wistar King A (WKA, RT-1k) splenocytes were established by the persistence of donor dendritic cells (DCs) in the host liver measured using fluorescence microscopy and flow cytometry and by the mixed lymphocyte reaction (MLR). The effect of intravenous gadolinium chloride (GDCl3) on the blockade of Kupffer cell function prior to portovenous administration of splenocytes was also assessed. The MLR response was strongly inhibited in a BN-restricted manner after portovenous administration of donor BN splenocytes, but not by venous nor by portovenous administration of WKA splenocytes. Immunosuppression was blocked by pretreatment with GDCl3. The percentage of donor DCs in hepatic non-parenchymal cells (NPCs) was significantly higher in the PV group compared with the IV group. Treatment with GDCl3 decreased the percentage of donor DCs. In addition, cytotoxic T lymphocyte antigen 4 (CTLA4/CD152), which may function as an immune attenuator, was strongly stained, and B7 was weakly stained in recipient liver in the PV group compared with the IV group. These results suggest that both donor DCs and recipient Kupffer cells (self DCs) are involved in the induction of immune hyporesponsiveness by donor cells. This occurs via portovenous administration, in which a signal of the CTLA4-B7 pathway played an important part in inhibiting the interaction of CD28 and its B7 ligands.
Collapse
Affiliation(s)
- K Nakagawa
- First Department of Surgery, Okayama University Medical School, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
|
32
|
Seino KI, Fukao K, Muramoto K, Yanagisawa K, Takada Y, Kakuta S, Iwakura Y, Van Kaer L, Takeda K, Nakayama T, Taniguchi M, Bashuda H, Yagita H, Okumura K. Requirement for natural killer T (NKT) cells in the induction of allograft tolerance. Proc Natl Acad Sci U S A 2001; 98:2577-81. [PMID: 11226281 PMCID: PMC30180 DOI: 10.1073/pnas.041608298] [Citation(s) in RCA: 205] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
In this study, we investigated the role of Valpha14 natural killer T (NKT) cells in transplant immunity. The ability to reject allografts was not significantly different between wild-type (WT) and Valpha14 NKT cell-deficient mice. However, in models in which tolerance was induced against cardiac allografts by blockade of lymphocyte function-associated antigen-1/intercellular adhesion molecule-1 or CD28/B7 interactions, long-term acceptance of the grafts was observed only in WT but not Valpha14 NKT cell-deficient mice. Adoptive transfer with Valpha14 NKT cells restored long-term acceptance of allografts in Valpha14 NKT cell-deficient mice. The critical role of Valpha14 NKT cells to mediate immunosuppression was also observed in vitro in mixed lymphocyte cultures in which lymphocyte function-associated antigen-1/intercellular adhesion molecule-1 or CD28/B7 interactions were blocked. Experiments using IL-4- or IFN-gamma-deficient mice suggested a critical contribution of IFN-gamma to the Valpha14 NKT cell-mediated allograft acceptance in vivo. These results indicate a critical contribution of Valpha14 NKT cells to the induction of allograft tolerance and provide a useful model to investigate the regulatory role of Valpha14 NKT cells in various immune responses.
Collapse
Affiliation(s)
- K I Seino
- Department of Surgery, Institute of Clinical Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba Science City, Ibaraki 305-8575, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Bachetoni A, D'Ambrosio A, Mariani P, Cortesini R, Quintieri F. Diltiazem affects human dendritic cell maturation. Transplant Proc 2001; 33:231-3. [PMID: 11266794 DOI: 10.1016/s0041-1345(00)01989-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- A Bachetoni
- II Clinica Chirurgica, Policlinico Umberto I, Università "La Sapienza," Rome, Italy
| | | | | | | | | |
Collapse
|
34
|
Xia G, Goebels J, Rutgeerts O, Vandeputte M, Waer M. Transplantation tolerance and autoimmunity after xenogeneic thymus transplantation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 166:1843-54. [PMID: 11160231 DOI: 10.4049/jimmunol.166.3.1843] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Successful grafting of vascularized xenografts (Xgs) depends on the ability to reliably induce both T cell-independent and -dependent immune tolerance. After temporary NK cell depletion, B cell suppression, and pretransplant infusion of donor Ags, athymic rats simultaneously transplanted with hamster heart and thymus Xgs developed immunocompetent rat-derived T cells that tolerated the hamster Xgs but provoked multiple-organ autoimmunity. The autoimmune syndrome was probably due to an insufficient development of tolerance for some rat organs; for example, it led to thyroiditis in the recipient rat thyroid, but not in simultaneously transplanted donor hamster thyroid. Moreover, grafting a mixed hamster/rat thymic epithelial cell graft could prevent the autoimmune syndrome. These experiments indicate that host-type thymic epithelial cells may be essential for the establishment of complete self-tolerance and that mixed host/donor thymus grafts may induce T cell xenotolerance while maintaining self-tolerance in the recipient.
Collapse
Affiliation(s)
- G Xia
- Laboratory for Experimental Transplantation, University of Leuven, Leuven, Belgium
| | | | | | | | | |
Collapse
|
35
|
Rogers SA, Liapis H, Hammerman MR. Transplantation of metanephroi across the major histocompatibility complex in rats. Am J Physiol Regul Integr Comp Physiol 2001; 280:R132-6. [PMID: 11124143 DOI: 10.1152/ajpregu.2001.280.1.r132] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
To determine whether transplanted metanephroi grow, differentiate, and function in hosts that differ in major histocompatibility complex loci (RT1 loci in rats) from donors in a defined way, we implanted metanephroi from embryonic day (E) 15 PVG (RT1(c)) rat embryos into the omentum of nonimmunosupressed uninephrectomized PVG-RT1(avl) (host) rats. By 4 wk posttransplantation, metanephroi had grown and differentiated such that glomeruli, proximal and distal tubules, and collecting ducts had normal structure and ultrastructure. At 12 wk posttransplantation, weights of metanephroi were 54 +/- 8 mg. Inulin clearances were 0.9 +/- 0.3 microl. min(-1). 100 g rat wt(-1). In vitro, splenocytes from PVG rats stimulated the proliferation of cells originating from both PVG-RT1(avl) rats in which a transplant had been performed and PVG-RT1(avl) rats with no transplant. Full-thickness PVG-RT1(avl) skin engrafted normally on PVG-RT1(avl) rats in which PVG metanephroi had been previously implanted and metanephroi retained a normal appearance. In contrast, skin from PVG rats sloughed, and the tubular architecture of metanephroi was obliterated by a mononuclear cell infiltrate consistent with acute rejection. Here we show for the first time that functional chimeric kidneys develop from metanephroi transplanted across the MHC into nonimmunosupressed hosts and provide evidence that a state of peripheral immune tolerance secondary to T cell "ignorance" permits their survival.
Collapse
Affiliation(s)
- S A Rogers
- George M. O'Brien Kidney and Urological Disease Center, Renal Division, Departments of Medicine, Cell Biology and Physiology, and Pathology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | | | | |
Collapse
|
36
|
Abstract
New drugs have recently been added that may eventually replace the two-decade dominance of cyclosporin in solid organ transplantation. This cornerstone of immunosuppression was introduced by Borel [1] and Calne [2] in the mid-70s. In 1989, Starzl et al., after 2 years of preclinical experimentation, introduced tacrolimus (originally designated as FK506 by the Fujisawa Pharmaceutical Company of Japan) as a potent immunosuppressant for liver transplants [3]. Also, in recent years, a variety of novel purine and pyrimidine biosynthesis inhibitors have been tested for transplantation therapy. The leading agent which appears to be replacing the 35-year position occupied by azathioprine is the semi-synthetic morpholinoethyl ester of mycophenolic acid (MPA), mycophenolate mofetil (MMF), introduced by Allison [4] and Sollinger [5], and developed by the Syntex Corporation (now Roche Pharmaceuticals). Others, affecting different intra- or intercellular messages amplifying immunity, are in the pipeline.
Collapse
Affiliation(s)
- G Ciancio
- Department of Surgery, Division of Transplantation, University of Miami School of Medicine, PO Box 012440, Miami, Fl. 33101, USA.
| | | | | |
Collapse
|
37
|
Shizuru JA, Weissman IL, Kernoff R, Masek M, Scheffold YC. Purified hematopoietic stem cell grafts induce tolerance to alloantigens and can mediate positive and negative T cell selection. Proc Natl Acad Sci U S A 2000; 97:9555-60. [PMID: 10920206 PMCID: PMC16903 DOI: 10.1073/pnas.170279297] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2000] [Indexed: 11/18/2022] Open
Abstract
Engraftment of allogeneic bone marrow (BM) has been shown to induce tolerance to organs genotypically matched with the BM donor. Immune reconstitution after BM transplantation therefore involves re-establishment of a T cell pool tolerant to antigens present on both donor and host tissues. However, how hematopoietic grafts exert their influence over the regenerating immune system is not completely understood. Prior studies suggest that education of the newly arising T cell pool involves distinct contributions from donor and host stromal elements. Specifically, negative selection is thought to be mediated primarily by donor BM-derived antigen-presenting cells, whereas positive selection is dictated by radio-resistant host-derived thymic stromal cells. In this report we studied the effect of highly purified allogeneic hematopoietic stem cells (HSCs) on organ transplantation tolerance induction and immune reconstitution. In contrast to engraftment of BM that results in near-complete donor T cell chimerism, HSC engraftment results in mixed T cell chimerism. Nonetheless we observed that HSC grafts induce tolerance to donor-matched neonatal heart grafts, and one way the HSC grafts alter host immune responses is via deletion of newly arising donor as well as radiation-resistant host T cells. Furthermore, using an in vivo assay of graft rejection to study positive selection we made the unexpected observation that T cells in chimeric mice rejected grafts only in the context of the donor MHC type. These latter findings conflict with the conventionally held view that radio-resistant host elements primarily dictate positive selection.
Collapse
Affiliation(s)
- J A Shizuru
- Departments of Medicine and Pathology, Stanford University School of Medicine, CA 94305, USA.
| | | | | | | | | |
Collapse
|
38
|
Vendetti S, Chai JG, Dyson J, Simpson E, Lombardi G, Lechler R. Anergic T cells inhibit the antigen-presenting function of dendritic cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 165:1175-81. [PMID: 10903714 DOI: 10.4049/jimmunol.165.3.1175] [Citation(s) in RCA: 134] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The phenomena of infectious tolerance and linked-suppression are well established, but the mechanisms involved are incompletely defined. Anergic T cells can inhibit responsive T cells in vitro and prolong skin allograft survival in vivo. In this study the mechanisms underlying these events were explored. Allospecific mouse T cell clones rendered unresponsive in vitro inhibited proliferation by responsive T cells specific for the same alloantigens. The inhibition required the presence of APC, in that the response to coimmobilized anti-CD3 and anti-CD28 Abs was not inhibited. Coculture of anergic T cells with bone marrow-derived dendritic cells (DC) led to profound inhibition of the ability of the DC to stimulate T cells with the same or a different specificity. After coculture with anergic T cells expression of MHC class II, CD80 and CD86 by DC were down-regulated. These effects did not appear to be due to a soluble factor in that inhibition was not seen in Transwell experiments, and was not reversed by addition of neutralizing anti-IL-4, anti-IL-10, and anti-TGF-beta Abs. Taken together, these data suggest that anergic T cells function as suppressor cells by inhibiting Ag presentation by DC via a cell contact-dependent mechanism.
Collapse
Affiliation(s)
- S Vendetti
- Department of Immunology, Imperial College School of Medicine, and Medical Research Council Clinical Sciences Centre, Imperial College School of Medicine, Hammersmith Hospital, London, United Kingdom
| | | | | | | | | | | |
Collapse
|
39
|
Kenyon NS, Chatzipetrou M, Tzakis A, Miller J, Alejandro R, Ricordi C. Allogeneic hematopoietic stem cell transplantation in recipients of cellular or solid organ allografts. Cancer Treat Res 2000; 101:109-32. [PMID: 10800647 DOI: 10.1007/978-1-4615-4987-1_6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- N S Kenyon
- Department of Medicine, University of Miami School of Medicine, FL, USA
| | | | | | | | | | | |
Collapse
|
40
|
|
41
|
Han D, Ricordi C, Xu X, Kenyon NS. Quantitative polymerase chain reaction assessment of chimerism in non-human primates after sex-mismatched islet and bone marrow transplantation. Transplantation 2000; 69:1717-21. [PMID: 10836386 DOI: 10.1097/00007890-200004270-00031] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
BACKGROUND Accurate assessment of chimerism in recipients of islet and bone marrow transplantation (BMT) may allow for a clearer assessment of the role of chimerism in islet engraftment or rejection. A quantitative polymerase chain reaction (PCR) assay was developed for the detection of the sex-determining region of the Y chromosome (SRY) in peripheral blood samples from female non-human primate recipients of allogeneic male islets and vertebral body marrow (VBM) from the same donor. METHODS The assay incorporates a synthetic internal standard (IS) containing the same primer template sequences as the target to compete for primer annealing and amplification. Each DNA sample was coamplified with a constant amount of IS. The concentration of male DNA in the test samples was calculated from the regression equation of a standard curve that was generated by plotting the logarithm of the ratio of the intensities of SRY to IS PCR products versus the logarithm of known percentages of input male DNA. RESULTS This method allows for a correction of the variability of efficiency of the PCR technique and also overcomes the drawback of time-consuming competitive PCR. Using this assay, we quantitated the amount of male DNA in samples taken from female baboon recipients of male islets and VBM. There was detectable male donor DNA in the samples taken one day after BMT; pre-BMT samples were negative. This technique works well for samples obtained from rhesus and cynomogus monkeys as well. CONCLUSIONS It is a practical method for accurately evaluation of chimerism after sex-mismatched allogeneic BMT in non-human primate models.
Collapse
Affiliation(s)
- D Han
- Diabetes Research Institute, University of Miami School of Medicine, FL 33136, USA
| | | | | | | |
Collapse
|
42
|
Abstract
The induction of tolerance to transplanted organs could make transplantation safer and more uniformly successful. One of the most promising approaches currently being investigated involves the induction of deletional tolerance through the establishment of "mixed chimerism." In this laboratory, we first studied mixed chimerism as an approach to transplantation tolerance in mice, using a nonmyeloablative preparative regimen consisting of 300 R whole-body irradiation, 700 R thymic irradiation, and treatment with monoclonal antibodies to CD4 and CD8. This approach has subsequently been extended successfully to the induction of tolerance to renal transplants in fully mismatched cynomolgus monkeys. In addition, the same approach, with minor modifications, has been found effective in producing mixed chimerism and transplantation tolerance in the concordant xenogeneic baboon to cynomolgus monkey species combination. Because pigs have many advantages as a potential xenograft donor for humans, we are also trying to extend our nonmyeloablative regimen for production of mixed chimerism to the discordant pig --> primate combination. We have used absorption of natural antibodies to prevent hyperacute rejection and then proceeded with a mixed chimerism approach. Administration of pig hematopoietic stem cells along with pig recombinant cytokines (SCF and IL-3) to primates has enabled the pig bone marrow to survive in these xenogeneic hosts for over 6 months. This chimerism has apparently been sufficient to markedly diminish T cell immunity and the induction of new T-cell-dependent responses. However, to date we have not succeeded in preventing the return of natural antibodies, which appear to be the cause of eventual loss of organ transplants and are the subject of further intensive investigations.
Collapse
Affiliation(s)
- D H Sachs
- Transplantation Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02129, USA
| |
Collapse
|
43
|
Gassel HJ, Otto C, Gassel AM, Meyer D, Steger U, Timmermann W, Ulrichs K, Thiede A. Tolerance of rat liver allografts induced by short-term selective immunosuppression combining monoclonal antibodies directed against CD25 and CD54 with subtherapeutic cyclosporine. Transplantation 2000; 69:1058-67. [PMID: 10762208 DOI: 10.1097/00007890-200003270-00008] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
BACKGROUND Our purpose was to develop and evaluate protocols for selective immunosuppression after liver transplantation using the monoclonal antibodies (mAbs) NDS-61, directed against the interleukin-2 receptor (CD25), and 1A29, directed against the intercellular adhesion molecule-1 (CD54), in combination with subtherapeutic cyclosporine (CsA). METHODS Orthotopic rat liver transplantation (ORLT) was performed in a DA-to-LEW strain combination. Immunosuppression was administered from day 0 to +13. Functional parameters such as survival time, body weight, and serum bilirubin levels were measured and the liver grafts were evaluated histologically. RESULTS A stepwise tapering of CsA from 3 to 0.25 mg/kg/day reduced the long-term survival rate. All animals died at a CsA dosage of 0.25 mg/kg/day, which was therefore defined as subtherapeutic. Monotherapy with the anti-CD25 mAb was performed at dosages of 600 and 1800 microg/kg/day. The lower mAb dosage resulted in a long-term survival rate of 12% and was defined as subtherapeutic. The combination therapy of CsA (0.25 mg/kg/day) and anti-CD25 mAb (600 microg/kg/day) produced a synergistic effect and led to a long-term survival rate of 84%. This survival rate was significantly higher than those after either CsA (P<0.005) or anti-CD25 mAb (P<0.001) monotherapy. Both dosages (10 and 30 microg/kg/day) of anti-CD54 mAb monotherapy as well as anti-CD54 mAb combined with a subtherapeutic dosage of CsA were ineffective in preventing acute allograft rejection. The addition of anti-CD54 mAb (30 microg/kg/day) to combined CsA plus anti-CD25 mAb therapy (triple therapy), however, increased the long-term survival rate to 100%. In the triple therapy group there was no rejection process in the liver allografts at any time, and donor-specific tolerance could be shown by donor-specific and third-party heterotopic heart transplantation. CONCLUSIONS The synergistic action of subtherapeutic CsA plus anti-CD25 mAb NDS-60 could be demonstrated, whereas anti-CD54 mAb only had a positive effect in a triple therapy group. Triple therapy prevented both acute and chronic rejection and induced donor-specific tolerance.
Collapse
Affiliation(s)
- H J Gassel
- Department of Surgery, University of Wuerzburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Xia GL, Ji P, Rutgeerts O, Waer M. Approaches toward T cell-independent and -dependent xenograft tolerance. Transplant Proc 2000; 32:371-3. [PMID: 10715443 DOI: 10.1016/s0041-1345(99)00983-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- G L Xia
- Laboratory for Experimental Transplantation, University of Leuven, Leuven, Belgium
| | | | | | | |
Collapse
|
45
|
Affiliation(s)
- M R Hammerman
- Washington University School of Medicine, St. Louis, Missouri 63110-1093, USA
| |
Collapse
|
46
|
Nicolaidou E, Okada Y, Zuo XJ, Toyoda M, Marchevsky A, Matloff J, Jordan SC. Prolongation of skin allograft survival is associated with reduced Th1 cytokine responses in the WKY-->F344 rat model. Transplantation 1999; 68:1393-401. [PMID: 10573081 DOI: 10.1097/00007890-199911150-00028] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
BACKGROUND We have reported previously that F344 rats develop a spontaneous tolerance to WKY lung allografts and show long-term retention of donor-specific skin grafts placed 35 days after lung transplantation. In this study, we investigated the immunologic mechanisms that may be responsible for the prolonged skin graft survival in animals tolerized with lung allografts. METHODS In the rejection group, WKY skin grafts were placed on normal F344 rats, whereas, in the tolerance group, the skin grafts were placed on F344 rats that had received a WKY lung transplant 35 days before skin grafting. Th1 (interleukin [IL]-2 and interferon-gamma [IFN-gamma]) and Th2 (IL-4 and IL-10) cytokine as well as transforming growth factor-beta1 mRNA expression in skin grafts and in draining lymph nodes were determined by reverse transcription-polymerase chain reaction. Macrophage and lymphocyte infiltration in skin grafts and the number of Langerhans cells in epidermal sheets of the grafts were examined by immunohistochemistry. RESULTS IL-2 and IFN-gamma mRNA expression was significantly decreased in both the skin grafts and the draining lymph nodes of the tolerance group, compared to the rejection group, whereas IL-10 and transforming growth factor-beta1 mRNA expression was similar in both groups and IL-4 mRNA was rarely detected. Decreased and delayed CD8+, macrophage, and natural killer cell infiltration in the skin grafts from the tolerance group was also detected. Similar reduction in the number of Langerhans cells in the epidermis of the grafts from both groups was seen on day 1 after skin grafting, and thereafter the number remained stable in both groups. CONCLUSIONS Reduced expression of Th1 cytokines and decreased infiltration of CD8+ cells, macrophages, and natural killer cells in the skin grafts may be responsible for prolongation of skin graft survival in the tolerance group.
Collapse
Affiliation(s)
- E Nicolaidou
- Steven Spielberg Pediatric Research Center, Department of Pathology, Cedars-Sinai Medical Center, UCLA School of Medicine, Los Angeles, California 90048, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Douillard P, Cuturi MC, Brouard S, Josien R, Soulillou JP. T cell receptor repertoire usage in allotransplantation: an overview. Transplantation 1999; 68:913-21. [PMID: 10532525 DOI: 10.1097/00007890-199910150-00001] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Lymphocytes express antigen receptors that allow the immune system to specifically recognize antigens. In transplantation, T cells play a critical role in the rejection process, and different protocols inhibiting T cell-mediated alloreactivity efficiently achieve prolongation of allograft survival. T cells can interact with alloantigens by two ways, either by the "indirect" pathway that correspond to the physiological mechanism of T cell immune recognition, or through the "direct" pathway where they recognize alloantigens directly on the surface of donor cells. If some T cells are specifically activated in allorecognition, one should be able to indirectly detect this "selection" by analyzing the T cell receptor usage that could be biased and reflect the preferential amplification of alloreactive lymphocyte subsets. Nevertheless compared with disease states such as cancer or autoimmunity the T cell receptor repertoire is still largely uncharacterized. We review the current results available on T cell repertoire usage in transplantation studies involving humans or various animal models. The T cell receptor repertoire involved in transplantation (restricted or unrestricted) and the features potentially common to alloimmune responses will be discussed.
Collapse
Affiliation(s)
- P Douillard
- Institut de Transplantation et de Recherche en Transplantation, Institut National de la Sante et de la Recherche Medicale (INSERM U437), NANTES, France
| | | | | | | | | |
Collapse
|
48
|
Affiliation(s)
- T Wekerle
- Transplantation Biology Research Center, Massachusetts General Hospital/Harvard Medical School, Boston 02129, USA
| | | |
Collapse
|
49
|
Yang Z, Rostami S, Koeberlein B, Barker CF, Naji A. Cardiac allograft tolerance induced by intra-arterial infusion of recombinant adenoviral CTLA4Ig. Transplantation 1999; 67:1517-23. [PMID: 10401757 DOI: 10.1097/00007890-199906270-00004] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Systemic administration of soluble recombinant fusion protein of cytotoxic T lymphocyte antigen 4 (CTLA4Ig) induces blockade of the CD28/B7 costimulatory pathway and promotes survival of allogeneic and xenogeneic grafts. We tested the efficacy of local expression of CTLA4Ig gene in the myocardium, induced by transduction with a recombinant adenovirus encoding the CTLA4Ig gene, on the survival of rat cardiac allografts. METHODS The donor hearts were perfused ex vivo with recombinant adenovirus encoding CTLA4Ig cDNA (AdCTLA4Ig) via intra-aorta coronary artery before transplantation. The distribution and duration of CTLA4Ig transgene expression in the myocardium was assessed by reverse transcriptase polymerase chain reaction (RT-PCR) or in situ RT-PCR after transplantation. RESULTS In situ RT-PCR demonstrated abundant expression of CTLA4Ig transgene in the endo-myocardium of AdCTLA4Ig-perfused cardiac grafts. Lewis and Brown Norway cardiac allografts transduced with AdCTLA4Ig survived indefinitely in nonimmunosuppressed Wistar Furth recipients. However, donor-strain skin grafts were rejected by long-term recipients of cardiac allografts, which also triggered the rejection of the primary heart grafts. CONCLUSIONS A single ex vivo intra-aortic infusion of recombinant adenovirus encoding the CTLA4Ig gene induced efficient transduction of the endo-myocardium and promoted the permanent survival of cardiac allografts in nonimmunosuppressed hosts. Despite the beneficial effect of local immunosuppression on cardiac allograft survival, the strategy failed to promote a state of donor-specific peripheral tolerance.
Collapse
Affiliation(s)
- Z Yang
- Department of Surgery, University of Pennsylvania Medical Center, Philadelphia 19104-4283, USA
| | | | | | | | | |
Collapse
|
50
|
Yamada J, Yoshida M, Taylor AW, Streilein JW. Mice with Th2-Biased Immune Systems Accept Orthotopic Corneal Allografts Placed in “High Risk” Eyes. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.162.9.5247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
CD4+ T cells of the Th1 type play a central role in acute rejection of solid tissue grafts, including orthotopic corneal allografts. Th1 cells, which mediate delayed hypersensitivity, are the polar opposites of CD4+ Th2 cells, and the latter cells cross-regulate Th1 cells through the unique pattern of cytokines they secrete. As such, Th2 cells may have a useful role to play in preventing rejection of corneal allografts. To test this possibility, the immune systems of adult mice were biased toward Th2 responses by immunization with keyhole limpet hemocyanin plus IFA. When immunized subsequently with either OVA or allogeneic corneal tissue, these mice acquired Ag-specific primed T cells of the Th2 type. More important, allogeneic corneas grafted into neovascularized eyes of Th2-biased mice experienced significantly enhanced survival. To demonstrate that enhanced survival was promoted by donor-specific Th2 cells, lymphoid cells from keyhole limpet hemocyanin-immune mice bearing healthy corneal allografts suppressed orthotopic corneal allograft rejection when adoptively transferred into naive, syngeneic recipients. We conclude that acceptance of corneal allografts in neovascularized mouse eyes can be significantly enhanced by biasing the recipient immune system toward Th2 responses.
Collapse
Affiliation(s)
- Jun Yamada
- Schepens Eye Research Institute, Harvard Medical School, Boston, MA 02114
| | - Munenori Yoshida
- Schepens Eye Research Institute, Harvard Medical School, Boston, MA 02114
| | - Andrew W. Taylor
- Schepens Eye Research Institute, Harvard Medical School, Boston, MA 02114
| | - J. Wayne Streilein
- Schepens Eye Research Institute, Harvard Medical School, Boston, MA 02114
| |
Collapse
|