351
|
Abstract
Chronic allograft rejection is a slowly progressive, insidious process in which the host immune system continues to mount an immunological attack on a transplanted organ, ultimately resulting in the failure of the graft. To varying degrees, all solid organ grafts are at risk for chronic rejection and undergo a stereotypic process of injury and inflammation, eventually leading to parenchymal fibrosis. The clinical consequences of chronic rejection are particularly apparent in thoracic transplantation, where both patient and graft survival decline steadily over time and the opportunities for re-transplantation or long-term extracorporeal support are limited. A variety of antigen-dependent and antigen-independent factors are known to modulate the propensity for an organ to undergo chronic rejection. Recent clinical and laboratory research has suggested that distinct immunologic mechanisms may underlie the process of chronic rejection. Ultimately, strategies to induce long-term tolerance to alloantigens will be necessary to prevent chronic rejection and to abrogate the deleterious sequelae of chronic immunosuppression.
Collapse
Affiliation(s)
- James S Allan
- Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| | | |
Collapse
|
352
|
Wekerle T, Blaha P, Asari R, Schmid M, Kiss C, Roth E, Muhlbacher F. Tolerance Induction Through Mixed Chimerism. Eur Surg 2002. [DOI: 10.1046/j.1563-2563.2002.02020.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
353
|
Giovino MA, Down JD, Jackson JD, Sykes M, Monroy RL, White-Scharf ME. Porcine hematopoiesis on primate stroma in long-term cultures: enhanced growth with neutralizing tumor necrosis factor-alpha and tumor growth factor-beta antibodies. Transplantation 2002; 73:723-31. [PMID: 11907417 DOI: 10.1097/00007890-200203150-00011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Donor hematopoiesis is at a competitive disadvantage when bone marrow transplantation is across species barriers. This could present major limitations to xenogeneic stem cell transplantation as an approach to tolerance induction. An in vitro model of xenogeneic engraftment was established to identify inhibitors of porcine hematopoiesis in a primate environment. METHODS Porcine bone marrow cells (BMC), in the presence or absence of primate CD34+ positive cells, were cultured for 4-6 weeks on primate stroma with porcine cytokines. Cellularity and growth of colony-forming cells were indicators of hematopoietic growth. Effects of soluble factors were determined by using Transwell inserts to separate porcine cells from stroma. Neutralizing antibodies for human transforming growth factor-beta (TGF-beta) and tumor necrosis factor-alpha (TNF-alpha) were added to cultures. RESULTS Porcine hematopoiesis can be maintained in long-term cultures on primate stroma with pig cytokines. Adding BMC to the stroma below Transwell-containing porcine cells dramatically inhibited porcine hematopoiesis, showing an inhibitory role for soluble factors. Neutralizing antibodies against TNF-alpha or TGF-beta caused a modest enhancement of porcine hematopoiesis; however, the combination of both led to a substantial increase. Inhibitory effects of these cytokines were confirmed by adding TNF-alpha and TGF-beta to porcine cultures. CONCLUSIONS Porcine cells may be more sensitive to inhibitory effects of TNF-alpha and TGF-beta than primate cells and are at a disadvantage when in a primate environment. Potential implications of this observation are discussed in the context of establishing specific immune tolerance via mixed chimerism to facilitate xenotransplantation.
Collapse
|
354
|
New directions in T-cell signal transduction and transplantation tolerance. Curr Opin Organ Transplant 2002. [DOI: 10.1097/00075200-200203000-00005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
355
|
|
356
|
Sakai Y, Kim DK, Iwasa S, Liang J, Watanabe T, Onodera M, Nakauchi H. Bone marrow chimerism prevents atherosclerosis in arterial walls of mice deficient in apolipoprotein E. Atherosclerosis 2002; 161:27-34. [PMID: 11882314 DOI: 10.1016/s0021-9150(01)00614-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE apolipoprotein E (apoE) is a key regulator in cholesterol-rich lipoprotein metabolism. Inherited deficiency of this protein results in type III hyperlipoproteinemia in humans. ApoE, especially that derived from macrophages, can efficiently protect against development of atherosclerotic lesion. To use stem cell gene therapy or mini-transplant in treating abnormal lipid metabolism and preventing atherosclerosis, a minimal level of bone marrow chimerism must be determined. METHODS lethally irradiated apoE deficient mice (12-16 weeks of age) fed on normal chow were transplanted with normal bone marrow cells (C57BL/6.Ly5.1) mixed with those of apoE deficient mice (C57BL/6.Ly5.2) at various ratios. Plasma cholesterol levels were determined every 3 weeks for up to 42 weeks. Areas of atherosclerotic lesion in the aortas were quantified 6 months post-transplant. Plasma apoE was measured by Western blot analysis. RESULTS bone marrow transplantation (BMT) in apoE (-/-) mice resulted in a detectable level of plasma apoE as determined by Western blot analysis. The plasma cholesterol levels in mice with > or = 60% chimerism were normalized by 6 weeks post-transplant. Mice with < or = 40% chimerism showed significant reductions, but not normalization, in the plasma cholesterol levels even at 42 weeks posttransplant. However, atherosclerotic areas observed in 10%-chimeric mice were significantly smaller than those in control mice (P<0.01). Immunohistochemical studies in 10%-chimeric mice revealed foam cells derived from donor marrow (apoE (+/+)) and expressed immunoreactive apoE in the atherosclerotic lesion. The positive signals by Western blot analysis were represented in the plasma of up to 8% of the chimeric mice. CONCLUSION chimerism of 10%, the minimum level analyzed, was sufficient to reduce the severity of atherosclerosis, although the plasma cholesterol levels were not completely normalized. The results indicate that stem cell gene therapy and mini-transplant may provide possible therapeutic approaches to treat patients with abnormal lipid metabolism and atherosclerosis.
Collapse
Affiliation(s)
- Yutaka Sakai
- Department of Immunology, Institute of Basic Medical Sciences, University of Tukuba, and CREST (Japan Science and Technology Corporation), 1-1-1 Ten-nodai, Tsukuba Science City, Ibaraki, 305-8575, Japan
| | | | | | | | | | | | | |
Collapse
|
357
|
Zhang QW, Tomita Y, Matsuzaki G, Shimizu I, Iwai T, Okano S, Minagawa R, Nakashima Y, Sueishi K, Nomoto K, Yasui H. Heart allograft tolerance without development of posttransplant cardiac allograft vasculopathy in chimerism-based, drug-induced tolerance. Transplantation 2002; 73:652-6. [PMID: 11889448 DOI: 10.1097/00007890-200202270-00031] [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: 11/26/2022]
Abstract
BACKGROUND Recently, we have described a drug (cyclophosphamide [CP] plus busulfan [BU])-induced skin allograft tolerance in mice that can regularly overcome fully H-2-mismatched barriers. Using this method, we have investigated whether or not this regimen can prolong the survival of heart allografts and inhibit the development of posttransplant cardiac allograft vasculopathy (CAV). METHODS The components of the method are intravenous administration of 1 x 108 allogeneic spleen cells on day 0, intraperitoneal injection of 200 mg/kg of CP and 30 mg/kg of BU on day 2, and intravenous injection of T cell-depleted 1 x 107 allogeneic bone marrow cells from the same strain of mice on day 3. Heart grafting was performed on day 28. Chimerism in peripheral blood was followed by flow cytometric analysis, and histological analysis was performed at various times after grafting. RESULTS In a fully major histocompatability complex (MHC)-mismatched combination of B10.D2 (H-2d, IE+)-->B10 (H-2b, IE-), stable, multilineage-mixed chimerism was observed permanently. B10.D2 heart grafts were accepted permanently in a donor-specific manner, and posttransplant CAV did not develop. CONCLUSIONS These results demonstrated that the drug-induced tolerance recently established by us can regularly induce a long-lasting heart allograft tolerance without development of CAV.
Collapse
Affiliation(s)
- Qi-Wei Zhang
- Department of Cardiovascular Surgery, Faculty of Medicine, Kyushu University, Fukuoka 812-8582, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
358
|
Fändrich F, Lin X, Chai GX, Schulze M, Ganten D, Bader M, Holle J, Huang DS, Parwaresch R, Zavazava N, Binas B. Preimplantation-stage stem cells induce long-term allogeneic graft acceptance without supplementary host conditioning. Nat Med 2002; 8:171-8. [PMID: 11821902 DOI: 10.1038/nm0202-171] [Citation(s) in RCA: 253] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Hematopoietic stem cells have been successfully employed for tolerance induction in a variety of rodent and large animal studies. However, clinical transplantation of fully allogeneic bone marrow or blood-borne stem cells is still associated with major obstacles, such as graft-versus-host disease or cytoreductive conditioning-related toxicity. Here we show that when rat embryonic stem cell-like cells of WKY origin are injected intraportally into fully MHC-mismatched DA rats, they engraft permanently (>150 days) without supplementary host conditioning. This deviation of a potentially alloreactive immune response sets the basis for long-term graft acceptance of second-set transplanted WKY cardiac allografts. Graft survival was strictly correlated with a state of mixed chimerism, which required functional thymic host competence. Our results provide a rationale for using preimplantation-stage stem cells as vehicles in gene therapy and for the induction of long-term graft acceptance.
Collapse
Affiliation(s)
- Fred Fändrich
- Department of General Surgery and Thoracic Surgery, University of Kiel, 24105 Kiel, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
359
|
Yamada A, Salama AD, Sayegh MH. The role of novel T cell costimulatory pathways in autoimmunity and transplantation. J Am Soc Nephrol 2002; 13:559-575. [PMID: 11805188 DOI: 10.1681/asn.v132559] [Citation(s) in RCA: 127] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Affiliation(s)
- Akira Yamada
- Laboratory of Immunogenetics and Transplantation, Renal Division, Brigham and Women's Hospital; Division of Nephrology, Children's Hospital; Harvard Medical School, Boston, Massachusetts
| | - Alan D Salama
- Laboratory of Immunogenetics and Transplantation, Renal Division, Brigham and Women's Hospital; Division of Nephrology, Children's Hospital; Harvard Medical School, Boston, Massachusetts
| | - Mohamed H Sayegh
- Laboratory of Immunogenetics and Transplantation, Renal Division, Brigham and Women's Hospital; Division of Nephrology, Children's Hospital; Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
360
|
Abstract
The creation of mixed hematopoietic chimerism has become an important clinical strategy for tolerance induction for cellular and organ transplantation, and for the treatment of numerous hematopoietic diseases. Clinical success has been limited however, by host immune response and by competition from host hematopoiesis. Recent data suggests that limited donor stem cell engraftment after minimally myeloablative hematopoietic stem cell (HSC) transplantation may in part be due to MHC associated microenvironmental mismatch resulting in a competitive disadvantage for donor HSC. A strategy to overcome this barrier to stable mixed hematopoietic chimerism would involve concurrent transplantation of a donor bone marrow microenvironment. To test this possibility, we set out to develop a method to tissue engineer a bone marrow microenvironment. One to two murine femurs were mechanically crushed to a fine suspension and were combined in vitro with various delivery vehicles. These constructs were transplanted into syngeneic animals in locations that are known to support transplantation of other tissues. Although bone formation was observed with several conditions, bone marrow formation was noted only within the small bowel mesentery when type I collagen was used as the delivery vehicle. No bone marrow formed when the vehicle was changed to polyglycolic acid or type IV collagen. We have demonstrated that the small bowel mesentery can support bone marrow formation under specific in vivo conditions. Future work will focus on strategies for transplantation of an engineered donor bone marrow environment to facilitate creation of allogeneic mixed hematopoietic chimerism.
Collapse
Affiliation(s)
- Alexander S Krupnick
- Children's Institute for Surgical Science, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104-4318, USA
| | | | | | | |
Collapse
|
361
|
Abstract
The CD154-CD40 pathway is one of the critical costimulatory pathways that are required for full activation of T cells during alloimmune responses. Blockade of this pathway with anti-CD154 antibodies has been reported to prolong allograft survival in experimental transplantation models and to induce tolerance in some instances. However, anti-CD154 monotherapy cannot induce tolerance in "stringent" models such as skin and islet transplantation and is not sufficient to prevent chronic graft vasculopathy in vascularized organ transplantation. Therefore, combined therapies of anti-CD154 antibodies plus donor-specific transfusion, bone marrow infusion, or B7 blockade by CTLA4-Ig have been tried, and synergistic effects for tolerance induction have been reported. Furthermore, the efficacy of CD154 blockade in primate models has been confirmed for islet and kidney transplantation. The mechanisms of CD154 blockade in vivo include CTLA4-dependent anergy or regulation, T-cell apoptosis, and induction of regulatory cells. Finally, anti-CD154 antibody therapy has been reported to result in unexpected thromboembolic complications in both primates and humans, although the etiology of these conditions remains unclear. In addition, not all antibodies cause this side effect. Clinical trials with humanized anti-CD154 monoclonal antibodies are underway in severe autoimmune diseases, but its development in transplantation is unclear at this time.
Collapse
Affiliation(s)
- Akira Yamada And
- Laboratory of Immunogenetics and Transplantation, Brigham and Women's Hospital, Transplantation Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | |
Collapse
|
362
|
Pierson RN, Crowe JE, Pfeiffer S, Atkinson J, Azimzadeh A, Miller GG. CD40-ligand in primate cardiac allograft and viral immunity. Immunol Res 2002; 23:253-62. [PMID: 11444390 DOI: 10.1385/ir:23:2-3:253] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Our laboratory has studied the role of CD40 ligand (CD40L, CD154) in the primate immune response to allogenic and infectious challenges. We find that intensive early blockade of CD40L reliably attenuates acute rejection of primate cardiac allografts. Monotherapy fails to prevent late graft loss, which often occurs in association with rising antidonor antibody titers and allograft vasculopathy, despite continuing anti-CD40L therapy. In contrast, the primary humoral response to T helper dependent influenza viral antigen is inhibited during anti-CD40L therapy, and responses to subsequent immunization are blunted after discontinuation of therapy. These results are encouraging with regard to the tolerogenic potential of costimulatory blockade for specific T helper dependent antigens. However, these findings also indicate that pathogenic allograft responses in primates are probably not entirely CD40L-dependent. As such, additional immunomodulatory strategies are needed to facilitate tolerance to a transplanted organ.
Collapse
MESH Headings
- Abatacept
- Animals
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Viral/biosynthesis
- Antibodies, Viral/immunology
- Antibody Specificity
- Antigens, CD
- Antigens, Differentiation/therapeutic use
- B-Lymphocytes/drug effects
- B-Lymphocytes/immunology
- CD40 Antigens/physiology
- CD40 Ligand/drug effects
- CD40 Ligand/physiology
- CTLA-4 Antigen
- Graft Enhancement, Immunologic
- Graft Rejection/immunology
- Graft Rejection/prevention & control
- Heart Transplantation/immunology
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Humans
- Immune Tolerance
- Immunoconjugates
- Isoantibodies/immunology
- Macaca fascicularis
- Mice
- Skin Transplantation/immunology
- T-Lymphocyte Subsets/immunology
- T-Lymphocytes, Helper-Inducer/immunology
- Time Factors
- Transplantation, Heterotopic
- Transplantation, Homologous
- Vaccination
Collapse
Affiliation(s)
- R N Pierson
- Vanderbilt University Medical Center, Department of Cardiothoracic Surgery, Nashville, TN, USA.
| | | | | | | | | | | |
Collapse
|
363
|
Nagahama T, Sugiura K, Lee S, Morita H, Adachi Y, Kwon AH, Kamiyama Y, Ikehara S. A new method for tolerance induction: busulfan administration followed by intravenous injection of neuraminidase-treated donor bone marrow. Stem Cells 2002; 19:425-35. [PMID: 11553851 DOI: 10.1634/stemcells.19-5-425] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The portal venous (p.v.) administration of foreign cells induces donor-specific tolerance. Recently, we have demonstrated that the p.v. administration of donor cells elicits donor-specific tolerance across major histocompatibility complex barriers. In the present study, utilizing the intrahepatic tolerance-inducing system, we have established a new method for organ transplantation using both busulfan ([Bu] to provide a sufficient "space" for the donor hematopoietic cells to expand in the recipient) and neuraminidase ([Neu] to enhance the trapping of i.v.-injected cells in the liver). Radiolabeled bone marrow cells (BMCs) were found to exclusively accumulate in the livers of the recipients as a result of the Neu treatment. Furthermore, hematopoietic progenitors (forming hematopoietic foci) in the accumulated BMCs were retained in the recipient livers for at least 18 days. C57BL/6 (B6) mice that had been transplanted with skins of BALB/c mice immediately after the injection of BALB/c BMCs showed a 90% skin graft survival rate over 400 days as a result of using the combination of injecting 50 mg/kg Bu into the B6 mice and treatment of the BALB/c BMCs with 0.25 U/ml Neu (50 Bu + 0.25 Neu). However, the survival rate significantly decreased when either the Bu or Neu treatment was omitted. In tolerant recipients, microchimerism was observed in the various hematolymphoid organs. T cells collected from the tolerant recipients suppressed proliferative responses to the donor-alloantigens but enhanced the production of Th2 and Th3 cytokines. These findings suggest that the enhanced retention of donor BMCs in the recipient livers as a result of the Bu and Neu treatments efficiently induces tolerance induction. Therefore, this "single-day protocol" would be of great advantage for human organ transplantation.
Collapse
Affiliation(s)
- T Nagahama
- The First Department of Pathology, Kansai Medical University, Osaka, Japan
| | | | | | | | | | | | | | | |
Collapse
|
364
|
Teshima T, Ferrara JLM. Understanding the alloresponse: new approaches to graft-versus-host disease prevention. Semin Hematol 2002; 39:15-22. [PMID: 11799525 DOI: 10.1053/shem.2002.29246] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Graft-versus-host disease (GVHD) has been the primary limitation to the wider application of allogeneic bone marrow transplantation (BMT). GVHD occurs when donor T cells react to host antigens on antigen-presenting cells (APCs) and attack host tissues, with sequential activation of donor T cells and monocytes/macrophages. The net effects of dysregulated cytokine production in this complex system are the severe inflammatory manifestations that we recognize as clinical acute GVHD. Long-term outcomes are also adversely affected by chronic GVHD, which has distinctive clinical and pathologic manifestations that mimic autoimmune disease, although its exact pathogenesis remains ambiguous. The ultimate goal for preventing GVHD is the induction of specific tolerance to host antigens, thereby maintaining favorable aspects of donor immunity. Tolerance may be achieved by costimulatory blockade, deletion of activated cells, suppression by regulatory T cells, and immune deviation. This report will focus on these mechanisms as they relate to the pathophysiology of acute GVHD.
Collapse
Affiliation(s)
- Takanori Teshima
- Department of Internal Medicine, University of Michigan Cancer Center, Ann Arbor, MI 48109-0942, USA
| | | |
Collapse
|
365
|
Adams AB, Larsen CP, Pearson TC, Newell KA. The role of TNF receptor and TNF superfamily molecules in organ transplantation. Am J Transplant 2002; 2:12-8. [PMID: 12095050 DOI: 10.1034/j.1600-6143.2002.020104.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The rapid increase in the number of molecules demonstrated to regulate immune responses has provided new opportunities for manipulation of the recipient immune response to transplanted organs. Molecules belonging to the TNF receptors and TNF superfamily are increasingly recognized as playing a major role in the regulation of immune responses to tumor, viral, and autoantigens. The mechanisms by which these molecules regulate immune responses are diverse. TNF receptor-related molecules have been shown to control the development of secondary lymphoid organs, affect the activation and survival of T cells and antigen presenting cells, and alter cytokine and chemokine production. An increasing amount of data suggest that some TNFR superfamily members are particularly important for the function of CD8+ T cells. Based on our current understanding of these molecules it seems highly likely that strategies that target selected TNFR/TNF superfamily molecules will be useful for controlling or preventing the rejection of transplanted organs and tissues.
Collapse
Affiliation(s)
- Andrew B Adams
- The Emory Transplant Center, Department of Surgery, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | |
Collapse
|
366
|
Shaw BE, Madrigal JA, Potter M. Improving the outcome of unrelated donor stem cell transplantation by molecular matching. Blood Rev 2001; 15:167-74. [PMID: 11792117 DOI: 10.1054/blre.2001.0163] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Volunteer unrelated donor (VUD) stem cell transplantation is now a well-established procedure in the treatment for many haematological and other disorders. The improved success of this modality of treatment is related, in part, to the existence of large volunteer donor registries (with well characterized tissue typing), as well as to the improved understanding of the molecular factors that have an influence on transplantation outcome. It is clear that close attention to human leukocyte antigen (HLA) matching is essential in ensuring a satisfactory transplant outcome, however the extent to which donor-recipient pairs need to be matched is not yet clear. There is also an increased understanding that factors other than HLA do affect clinical outcome. The ability to perform high resolution molecular typing techniques has allowed researchers to begin assessing the significance of mismatches at particular loci against an otherwise matched background, and in this way highlight the effects of individual genetic factors on transplantation outcome.
Collapse
Affiliation(s)
- B E Shaw
- Anthony Nolan Research Institute, Hampstead, UK.
| | | | | |
Collapse
|
367
|
Noris M, Cugini D, Casiraghi F, Azzollini N, Moraes LDDV, Mister M, Pezzotta A, Cavinato RA, Aiello S, Perico N, Remuzzi G. Thymic microchimerism correlates with the outcome of tolerance-inducing protocols for solid organ transplantation. J Am Soc Nephrol 2001; 12:2815-2826. [PMID: 11729252 DOI: 10.1681/asn.v12122815] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
This study found that pretransplant infusion of donor peripheral blood leukocytes, either total leukocytes (peripheral blood leukocytes) or peripheral blood mononuclear cells (PBMC), under appropriate immunomodulating conditions was more effective than donor bone marrow (BM) in prolonging the survival of rats that received kidney grafts. A higher percentage of MHCII(+) cells was found in donor PBMC than in BM cells, and depletion of MHCII(+) cells from donor PBMC abolished their tolerogenic potential. By the analysis of microchimerism in rats infused with donor cells and killed at different time points thereafter, the better tolerogenic potential of leukocyte infusion related to a higher capability of these cells to engraft the recipient thymus. PCR analysis on OX6-immunopurified cells revealed the presence of donor MHCII(+) cells in the thymus of these animals. The role of intrathymic microchimerism was reinforced by findings that thymectomy at the time of transplant prevented tolerance induction by donor leukocytes. Donor DNA was found in the thymus of most long-term graft animals that survived, but in none of those that rejected their grafts. The presence of intrathymic microchimerism correlated with graft survival, and microchimerism in other tissues was irrelevant. PCR analysis of DNA from thymic cell subpopulations revealed the presence of donor MHCII(+) cells in the thymus of long-term surviving animals. Thus, in rats, donor leukocyte infusion is better than donor BM for inducing graft tolerance, defined by long-term graft survival, donor-specific T cell hyporesponsiveness, and reduced interferon gamma production. This effect appears to occur through migration of donor MHCII(+) cells in the host thymus.
Collapse
Affiliation(s)
- Marina Noris
- Department of Immunology and Clinics of Organ Transplantation, Mario Negri Institute for Pharmacological Research, via Gavazzeni 11, 24125 Bergamo, Italy
| | - Daniela Cugini
- Department of Immunology and Clinics of Organ Transplantation, Mario Negri Institute for Pharmacological Research, via Gavazzeni 11, 24125 Bergamo, Italy
| | - Federica Casiraghi
- Department of Immunology and Clinics of Organ Transplantation, Mario Negri Institute for Pharmacological Research, via Gavazzeni 11, 24125 Bergamo, Italy
| | - Nadia Azzollini
- Department of Immunology and Clinics of Organ Transplantation, Mario Negri Institute for Pharmacological Research, via Gavazzeni 11, 24125 Bergamo, Italy
| | - Luciana De Deus Viera Moraes
- Department of Immunology and Clinics of Organ Transplantation, Mario Negri Institute for Pharmacological Research, via Gavazzeni 11, 24125 Bergamo, Italy
| | - Marilena Mister
- Department of Immunology and Clinics of Organ Transplantation, Mario Negri Institute for Pharmacological Research, via Gavazzeni 11, 24125 Bergamo, Italy
| | - Angela Pezzotta
- Department of Immunology and Clinics of Organ Transplantation, Mario Negri Institute for Pharmacological Research, via Gavazzeni 11, 24125 Bergamo, Italy
| | - Regiane Aparecida Cavinato
- Department of Immunology and Clinics of Organ Transplantation, Mario Negri Institute for Pharmacological Research, via Gavazzeni 11, 24125 Bergamo, Italy
| | - Sistiana Aiello
- Department of Immunology and Clinics of Organ Transplantation, Mario Negri Institute for Pharmacological Research, via Gavazzeni 11, 24125 Bergamo, Italy
| | - Norberto Perico
- Department of Immunology and Clinics of Organ Transplantation, Mario Negri Institute for Pharmacological Research, via Gavazzeni 11, 24125 Bergamo, Italy
| | - Giuseppe Remuzzi
- Department of Immunology and Clinics of Organ Transplantation, Mario Negri Institute for Pharmacological Research, via Gavazzeni 11, 24125 Bergamo, Italy
| |
Collapse
|
368
|
Shimoni A, Nagler A. Non-myeloablative stem cell transplantation (NST): chimerism testing as guidance for immune-therapeutic manipulations. Leukemia 2001; 15:1967-75. [PMID: 11753620 DOI: 10.1038/sj.leu.2402316] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2001] [Accepted: 07/19/2001] [Indexed: 11/08/2022]
Affiliation(s)
- A Shimoni
- Department of Bone Marrow Transplantation, Chaim Sheba Medical Center, Tel-Hashomer, Israel
| | | |
Collapse
|
369
|
Mehl AM, Jones M, Rowe M, Brennan P. Characterization of a CD40-dominant inhibitory receptor mutant. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:6388-93. [PMID: 11714804 DOI: 10.4049/jimmunol.167.11.6388] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
CD40 is an important mediator of immune and inflammatory responses. It is a costimulatory molecule for B cell proliferation and survival. Blockade of CD40 has been shown to induce tolerance and its role in other pathogenic conditions has led to the proposal that CD40 inhibition could be valuable therapeutically. As a first step to this end, we have characterized a CD40-dominant negative receptor. This inhibitory mutant lacks the identified CD40 signaling domains. It inhibits both cotransfected and endogenous CD40 activation of NF-kappaB. This mutant is specific, as it does not affect TNF or latent membrane protein 1 signaling. Its potential usefulness is illustrated by its ability to inhibit the CD40 ligand-stimulated increases of HLA and CD54 expression, molecules involved in Ag recognition and lymphocyte recruitment leading to organ rejection. The inhibitory mutant has no TNFR-associated factor 2-binding capabilities and inhibits the recruitment of TNFR-associated factor 2 to the CD40 signaling complex after stimulation. These studies show that the CD40 inhibitory receptor molecule is effective, specific, and useful both for research and potentially as a clinical tool. And furthermore, it is likely that similar dominant inhibitory receptors can be generated for all of the members of the TNFR superfamily.
Collapse
Affiliation(s)
- A M Mehl
- Infection and Immunity, University of Wales College of Medicine, Cardiff, United Kingdom
| | | | | | | |
Collapse
|
370
|
Kurtz J, Ito H, Wekerle T, Shaffer J, Sykes M. Mechanisms involved in the establishment of tolerance through costimulatory blockade and BMT: lack of requirement for CD40L-mediated signaling for tolerance or deletion of donor-reactive CD4+ cells. Am J Transplant 2001; 1:339-49. [PMID: 12099378 DOI: 10.1034/j.1600-6143.2001.10409.x] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
We have previously shown that high levels of multiline-age mixed hematopoietic chimerism and systemic T-cell tolerance can be achieved in mice without myeloablation through the use of anti-CD40L and costimulatory blockade alone (plus CTLA4Ig) or with recipient CD8 depletion and allogeneic bone marrow transplantation. Chimeric mice permanently accept donor skin grafts (> 100 days), and rapidly reject third-party grafts. The mechanisms by which costimulatory blockade facilitates the engraftment of allogeneic hematopoietic cells have not been defined. To further understand the in vivo mechanisms by which the administration of anti-CD40L mAb facilitates the engraftment of donor bone marrow and rapidly tolerizes CD4+ T cells, we analyzed the establishment of chimerism and tolerance in CD40L -/- mice. We demonstrate here that anti-CD40L mAb treatment is required only to prevent CD40L/CD40 interactions, and that no signal to the T cell through CD40L is necessary for the induction of CD4+ tolerance. Peripheral deletion of donor-reactive CD4+ T cells occurs rapidly in CD40L -/- mice receiving bone marrow transplantation (BMT), indicating that this deletion in the presence of anti-CD40L is not due to targeting of activated CD4+ cells by the antibody. Complete CD4+ cell tolerance is observed by both skin graft acceptance and in vitro assays before deletion is complete, indicating that additional mechanisms play a role in inducing CD4+ T-cell tolerance as the result of BMT in the presence of CD40/CD40L blockade.
Collapse
Affiliation(s)
- J Kurtz
- Transplantation Biology Research Center, Massachusetts General Hospital/Harvard Medical School, Boston 02129, USA
| | | | | | | | | |
Collapse
|
371
|
Williams MA, Tan JT, Adams AB, Durham MM, Shirasugi N, Whitmire JK, Harrington LE, Ahmed R, Pearson TC, Larsen CP. Characterization of virus-mediated inhibition of mixed chimerism and allospecific tolerance. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:4987-95. [PMID: 11673506 DOI: 10.4049/jimmunol.167.9.4987] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Simultaneous blockade of the CD28 and CD40 T cell costimulatory pathways has been shown to effectively promote skin allograft survival in mice. Furthermore, blockade of one or both of these pathways has played a central role in the development of strategies to induce mixed hematopoietic chimerism and allospecific tolerance. It has recently been observed that the beneficial effects of CD40 blockade and donor splenocytes in prolonging skin graft survival can be abrogated by some viral infections, including lymphocytic choriomeningitis virus (LCMV). In this study, we show that LCMV infection prevents prolonged allograft survival following CD28/CD40 combined blockade. We further show that LCMV prevents the induction of allospecific tolerance and mixed hematopoietic chimerism, while delay of infection for 3-4 wk posttransplant has no effect on tolerance induction. Because of reports of anti-H-2(d) activity following LCMV infection, we assayed the ability of LCMV-specific T cells to respond to alloantigen at a single cell level. Although we confirm that LCMV infection induces the generation of alloreactive cells, we also demonstrate that LCMV-specific T cells do not divide in response to alloantigen. The alloresponse suppressed by costimulation blockade is restored by LCMV infection and correlates with increased dendritic cell maturation. We hypothesize that the costimulation blockade-resistant rejection mediated by LCMV could be partly attributable to the up-regulation of alternative costimulatory pathways subsequent to LCMV-induced dendritic cell maturation.
Collapse
Affiliation(s)
- M A Williams
- Carlos and Marguerite Mason Transplantation Research Center and Department of Surgery, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
372
|
Ruggeri L, Capanni M, Martelli MF, Velardi A. Cellular therapy: exploiting NK cell alloreactivity in transplantation. Curr Opin Hematol 2001; 8:355-9. [PMID: 11604575 DOI: 10.1097/00062752-200111000-00007] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Allogeneic hematopoietic transplantation relies on T-cell alloreactions for engraftment and the graft-versus-leukemia (GVL) effect. In human leukocyte antigen (HLA) haplotype-mismatched transplants, extensive T-cell depletion of the graft is essential to prevent GVHD. This raises the question of whether mismatched transplants exert any GVL effect and whether it will ever be possible to reduce the intensity of preparative regimens. Because natural killer (NK) cells are negatively regulated by MHC class I-specific inhibitory receptors, mismatched transplants may trigger NK-cell alloreactivity. HLA class I disparities driving NK-cell alloreactions in the GVH direction mediate strong GVL effects, produce higher engraftment rates, and do not cause GVHD. In murine MHC-mismatched transplant models with no donor T-cell reactivity against the recipient, the pre-transplant infusion of donor-vs-recipient alloreactive NK cells conditioned the recipients to bone marrow transplantation without GVHD. NK-cell alloreactivity may be a unique therapeutic tool for tolerance induction and clearance of leukemia in hematopoietic transplantation.
Collapse
Affiliation(s)
- L Ruggeri
- Division of Hematology and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Perugia, Perugia, Italy
| | | | | | | |
Collapse
|
373
|
Basker M, Alwayn IP, Buhler L, Harper D, Abraham S, Kruger Gray H, DeAngelis H, Awwad M, Down J, Rieben R, White-Scharf ME, Sachs DH, Thall A, Cooper DK. Clearance of mobilized porcine peripheral blood progenitor cells is delayed by depletion of the phagocytic reticuloendothelial system in baboons. Transplantation 2001; 72:1278-85. [PMID: 11602856 DOI: 10.1097/00007890-200110150-00017] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
INTRODUCTION Attempts to achieve immunological tolerance to porcine tissues in nonhuman primates through establishment of mixed hematopoietic chimerism are hindered by the rapid clearance of mobilized porcine leukocytes, containing progenitor cells (pPBPCs), from the circulation. Eighteen hours after infusing 1-2 x 10(10) pPBPC/kg into baboons that had been depleted of circulating anti-alphaGal and complement, these cells are almost undetectable by flow cytometry. The aim of the present study was to identify mechanisms that contribute to rapid clearance of pPBPCs in the baboon. This was achieved by depleting, or blocking the Fc-receptors of, cells of the phagocytic reticuloendothelial system (RES) using medronate liposomes (MLs) or intravenous immunoglobulin (IVIg), respectively. METHODS Baboons (preliminary studies, n=4) were used in a dose-finding and toxicity study to assess the effect of MLs on macrophage depletion in vivo. In another study, baboons (n=9) received a nonmyeloablative conditioning regimen (NMCR) aimed at inducing immunological tolerance, including splenectomy, whole body irradiation (300 cGy) or cyclophosphamide (80 mg/kg), thymic irradiation (700 cGy), T-cell depletion, complement depletion with cobra venom factor, mycophenolate mofetil, anti-CD154 monoclonal antibody, and multiple extracorporeal immunoadsorptions of anti-alphaGal antibodies. The baboons were divided into three groups: Group 1 (n=5) NMCR+pPBPC transplantation; Group 2 (n=2) NMCR+ML+pPBPC transplantation; and Group 3 (n=2) NMCR+IVIg+pPBPC transplantation. Detection of pig cells in the blood was assessed by fluorescence-activated cell sorter and polymerase chain reaction (PCR). RESULTS PRELIMINARY STUDIES ML effectively depleted macrophages from the circulation in a dose-dependent manner. Group 1: On average, 14% pig cells were detected 2 hr postinfusion of 1 x 10(10) pPBPC/kg. After 18 hr, there were generally less than 1.5% pig cells detectable. Group 2: Substantially higher levels of pig cell chimerism (55-78%) were detected 2 hr postinfusion, even when a smaller number (0.5-1 x 10(10)/kg) of pPBPCs had been infused, and these levels were better sustained 18 hr later (10-52%). Group 3: In one baboon, 4.4% pig cells were detected 2 hr after infusion of 1 x 10(10) pPBPC/kg. After 18 hr, however, 7.4% pig cells were detected. A second baboon died 2 hr after infusion of 4 x 10(10) pPBPC/kg, with a total white blood cell count of 90,000, of which 70% were pig cells. No differences in microchimerism could be detected between the groups as determined by PCR. CONCLUSIONS This is the first study to report an efficient decrease of phagocytic function by depletion of macrophages with MLs in a large-animal model. Depletion of macrophages with MLs led to initial higher chimerism and prolonged the survival of circulating pig cells in baboons. Blockade of macrophage function with IVIg had a more modest effect. Cells of the RES, therefore, play a major role in clearing pPBPCs from the circulation in baboons. Depletion or blockade of the RES may contribute to achieving mixed hematopoietic chimerism and induction of tolerance to a discordant xenograft.
Collapse
Affiliation(s)
- M Basker
- Transplantation Biology Research Center, Massachusetts General Hospital / Harvard Medical School, Boston 02129, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
374
|
Melo ME, El-Amine M, Tonnetti L, Fleischman L, Scott DW. Gene therapeutic approaches to induction and maintenance of tolerance. Int Rev Immunol 2001; 20:627-45. [PMID: 11890616 DOI: 10.3109/08830180109045582] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Tolerance induction would be an ideal way to treat autoimmune diseases, especially if achievable in primed individuals. Moreover, specific tolerance would preclude the need for immunosuppressive treatment with its side effects. In this review, we will revisit the historical concepts of tolerance, and introduce a novel approach to tolerance via gene therapy. Our model system is based on the tolerogenicity of IgG carriers and B-cell antigen presentation. Results with this model demonstrate that it is simple and effective even in primed recipients, and has proven efficacy in three autoimmune models. We discuss the mechanisms of tolerance with fusion IgG's and analyze how our model of gene therapy approached can be utilized to fit in the future treatment of autoimmune conditions.
Collapse
Affiliation(s)
- M E Melo
- Dept of Immunology, American Red Cross, Jerome Holland Laboratory, Rockville, MD 20855, USA
| | | | | | | | | |
Collapse
|
375
|
Abstract
Developments in marrow and organ transplantation are mutually interactive. There have been several recent advances in stem cell transplantation: to ensure engraftment using larger doses of stem cells; to substantially reduce the incidence of graft-versus-host disease and marrow rejection using monoclonal antibodies; and to reduce toxicity of the preparative regimen through use of so-called nonmyeloablative regimens (mini-transplants). These advances may pave the way for generation of mixed hemopoietic chimerism as an aid to achieving tolerance to organ transplants. The use of short courses of T-cell-depleting antibodies, such as CD3 immunotoxin in primates and CAMPATH-1H in humans, has demonstrated that long-term graft survival may be possible without substantive long-term immunosuppressive treatment of the recipient. The demonstration in rodents that nondepleting antibodies to T cells can give rise to powerful regulatory mechanisms that maintain tolerance to grafts has initiated a major research effort in understanding how these regulatory T cells work, with the prospect of new therapeutic modalities to mimic or enhance their function.
Collapse
Affiliation(s)
- H Waldmann
- Sir William Dunn School of Pathology, South Parks Road, OX13RE, Oxford, UK.
| |
Collapse
|
376
|
Lee S, Sugiura K, Nagahama T, Iwai H, Yasumizu R, Yamashita T, Ikehara S. New method for thyroid transplantation across major histocompatibility complex barriers using allogeneic bone marrow transplantation. Transplantation 2001; 72:1144-9. [PMID: 11579314 DOI: 10.1097/00007890-200109270-00027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND It has been shown that allogeneic bone marrow transplantation (BMT) after lethal irradiation elicits donor-specific tolerance for organ or tissue transplantation across major histocompatibility complex (MHC) barriers. Recently, we have demonstrated that the portal venous (p.v.) administration of donor bone marrow cells (BMCs) elicits donor-specific tolerance across MHC barriers by only two administrations of an immunosuppressant (CsA or FK-506). In our study, using the central and intrahepatic tolerance-inducing system, we have established a new method for thyroid transplantation with BMT that would be more applicable to humans. METHODS In addition to sublethal (6-5 Gy) irradiation, recipient B6 (H-2b) mice received injections i.p. with the myeloablative drug busulfan (BU) on day -2 to provide a sufficient "space" for the donor hematopoietic cells to expand in the recipients. To induce the intrahepatic tolerance, donor BALB/c (H-2d) BMCs were treated with neuraminidase (Neu), which enhances the trapping of i.v. injected BMCs in the liver. After the injection of Neu-treated BMCs, the thyroid organs from the BALB/c mice were engrafted under the renal capsules. RESULTS A 90% graft survival rate was obtained over 100 days by a combination of BU administration, 6 Gy irradiation, and i.v. injection of Neu-treated BMCs [BU+6 Gy+(Neu) i.v.], and a 70% graft survival rate was obtained by [BU+5 Gy+(Neu) i.v.]. However, the graft survival rate significantly decreased when either the BU or Neu treatment was omitted. T cells collected from the tolerant recipients suppressed the proliferative responses to donor alloantigens. CONCLUSIONS Using both BU and Neu treatments, we have succeeded in inducing long-term tolerance and preventing the rejection of thyroid allografts by the single-day protocol.
Collapse
Affiliation(s)
- S Lee
- First Department of Pathology, Kansai Medical University, 10-15 Fumizono-cho, Moriguchi City, Osaka 570-8506, Japan
| | | | | | | | | | | | | |
Collapse
|
377
|
Wells AD, Li XC, Strom TB, Turka LA. The role of peripheral T-cell deletion in transplantation tolerance. Philos Trans R Soc Lond B Biol Sci 2001; 356:617-23. [PMID: 11375065 PMCID: PMC1088449 DOI: 10.1098/rstb.2001.0845] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The apoptotic deletion of thymocytes that express self-reactive antigen receptors is the basis of central (thymic) self-tolerance. However, it is clear that some autoreactive T cells escape deletion in the thymus and exist as mature lymphocytes in the periphery. Therefore, peripheral mechanisms of tolerance are also crucial, and failure of these peripheral mechanisms leads to autoimmunity. Clonal deletion, clonal anergy and immunoregulation and/or suppression have been suggested as mechanisms by which 'inappropriate' T-lymphocyte responses may be controlled in the periphery. Peripheral clonal deletion, which involves the apoptotic elimination of lymphocytes, is critical for T-cell homeostasis during normal immune responses, and is recognized as an important process by which self-tolerance is maintained. Transplantation of foreign tissue into an adult host represents a special case of 'inappropriate' T-cell reactivity that is subject to the same central and peripheral tolerance mechanisms that control reactivity against self. In this case, the unusually high frequency of naive T cells able to recognize and respond against non-self-allogeneic major histocompatibility complex (MHC) antigens leads to an exceptionally large pool of pathogenic effector lymphocytes that must be controlled if graft rejection is to be avoided. A great deal of effort has been directed toward understanding the role of clonal anergy and/or active immunoregulation in the induction of peripheral transplantation tolerance but, until recently, relatively little progress had been made towards defining the potential contribution of clonal deletion. Here, we outline recent data that define a clear requirement for deletion in the induction of peripheral transplantation tolerance across MHC barriers, and discuss the potential implications of these results in the context of current treatment modalities used in the clinical transplantation setting.
Collapse
Affiliation(s)
- A D Wells
- Department of Medicine, University of Pennsylvania, 415 Curie Boulevard, Philadelphia, PA 19104, USA
| | | | | | | |
Collapse
|
378
|
Kirk AD, Blair PJ, Tadaki DK, Xu H, Harlan DM. The role of CD154 in organ transplant rejection and acceptance. Philos Trans R Soc Lond B Biol Sci 2001; 356:691-702. [PMID: 11375072 PMCID: PMC1088456 DOI: 10.1098/rstb.2001.0855] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
CD154 plays a critical role in determining the outcome of a transplanted organ. This simple statement is amply supported by experimental evidence demonstrating that anti-CD154 antibodies are potent inhibitors of allograft rejection in many rigorous transplant models. Unfortunately, despite intensive investigation over the past ten years, the precise mechanisms by which antibodies against CD154 exert their anti-rejection effects have remained less obvious. Though originally classified with reference to B-cell function, CD154-CD40 interactions have also been shown to be important in T cell-antigen-presenting cell interactions. Accordingly, CD154 has been classified as a T-cell co-stimulatory molecule. However, mounting data suggest that treatment with anti-CD154 antibodies does not simply block costimulatory signals, but rather that the antibodies appear to induce signalling in receptor-bearing T cells. Other data suggest that anti-CD154 effects may be mediated by endothelial cells and possibly even platelets. In fact, the current literature suggests that CD154 can either stimulate or attenuate an immune response, depending upon the model system under study. CD154 has secured a fundamental place in transplant biology and general immunology that will no doubt be the source of considerable investigation and therapeutic manipulation in the coming decade.
Collapse
Affiliation(s)
- A D Kirk
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health/Navy, Naval Medical Research Center, Building 10, Room 11S/219, Center Drive, Bethesda, MD 20892, USA.
| | | | | | | | | |
Collapse
|
379
|
Adams AB, Pearson TC, Larsen CP. Conventional immunosuppression and co-stimulation blockade. Philos Trans R Soc Lond B Biol Sci 2001; 356:703-5. [PMID: 11375073 PMCID: PMC1088457 DOI: 10.1098/rstb.2001.0854] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Organ transplantation has become an accepted and successful therapeutic intervention for many patients with end-stage organ disease. Current conventional immunosuppressive regimens achieve very good short-term allograft survival but long-term outcomes are less than adequate. Furthermore, non-specific immunosuppression has its attendant side-effects including increased risks of malignancy and infection as well as drug-specific sequellae. With recent advances in the field of immunology, promising new therapies have arisen that could potentially eliminate lifelong drug therapy and promote indefinite acceptance of the donor tissue. Identification of co-stimulatory signals essential for T-cell activation has provided exciting new possibilities for controlling the alloimmune response. The compatibility of these new agents with proven conventional therapeutics has yielded mixed results. When used in combination, their immunosuppressive properties appear synergistic. However, if the goal of therapy is sustained, specific T-cell hyporesponsiveness, many conventional agents antagonize the effects of co-stimulatory blockade in several immune tolerance models.
Collapse
Affiliation(s)
- A B Adams
- The Carlos and Marguerite Mason Transplantation Research Center, Department of Surgery, Emory University School of Medicine, 5105 Woodruff Memorial Research Building, 1639 Pierce Drive, Atlanta, GA 30322, USA
| | | | | |
Collapse
|
380
|
Inverardi L, Ricordi C. Tolerance and pancreatic islet transplantation. Philos Trans R Soc Lond B Biol Sci 2001; 356:759-65. [PMID: 11375078 PMCID: PMC1088462 DOI: 10.1098/rstb.2001.0849] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Islet transplantation holds renewed promise as a cure for type I diabetes mellitus. Results of recent clinical trials have shown remarkable success, and have reignited universal optimism for this procedure. In spite of this success, the need for life-long immunosuppression of the recipient still limits islet transplantation to patients with poorly controlled diabetes or to those requiring kidney transplantation. It is obvious that the achievement of immunological tolerance would broaden the indication for islet transplantation to a much larger cohort of patients with type I diabetes mellitus, most likely preventing long-term complications and contributing to a much improved quality of life. Increased understanding of the basic mechanisms of tolerance induction has resulted in the implementation of numerous experimental approaches to achieve long-term survival of islet grafts in the absence of chronic immunosuppression. In this brief review we will attempt to summarize the current status of research and knowledge.
Collapse
Affiliation(s)
- L Inverardi
- Diabetes Research Institute, Cell Transplant Center, University of Miami School of Medicine, 1450 NW 10th Avenue, Miami, FL 33136, USA
| | | |
Collapse
|
381
|
Field EH, Strober S. Tolerance, mixed chimerism and protection against graft-versus-host disease after total lymphoid irradiation. Philos Trans R Soc Lond B Biol Sci 2001; 356:739-48. [PMID: 11375076 PMCID: PMC1088460 DOI: 10.1098/rstb.2001.0851] [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: 11/12/2022] Open
Abstract
Total lymphoid irradiation (TLI), originally developed as a non-myeloablative treatment for Hodgkin's disease, has been adapted for the induction of immune tolerance to organ allografts in rodents, dogs and non-human primates. Moreover, pretransplantation TLI has been used in prospective studies to demonstrate the feasibility of the induction of tolerance to cadaveric kidney allografts in humans. Two types of tolerance, chimeric and non-chimeric, develop after TLI treatment of hosts depending on whether donor bone marrow cells are transplanted along with the organ allograft. An advantageous feature of TLI for combined marrow and organ transplantation is the protection against graft-versus-host disease (GVHD) and facilitation of chimerism afforded by the predominance of CD4+ NK1.1(+) -like T cells in the irradiated host lymphoid tissues. Recently, a completely post-transplantation TLI regimen has been developed resulting in stable mixed chimerism and tolerance that is enhanced by a brief course of cyclosporine. The post-transplantation protocol is suitable for clinical cadaveric kidney transplantation. This review summarizes the evolution of TLI protocols for eventual application to human clinical transplantation and discusses the mechanisms involved in the induction of mixed chimerism and protection from GVHD.
Collapse
Affiliation(s)
- E H Field
- Department of Veterans Affairs Medical Center, Iowa City, IA 52246, USA
| | | |
Collapse
|
382
|
Abstract
Induction of mixed chimerism has the potential to overcome the current limitations of transplantation, namely chronic rejection, complications of immunosuppressive therapy and the need for xenografts to overcome the current shortage of allogeneic organs. Successful achievement of mixed chimerism had been shown to tolerize T cells, B cells and possibly natural killer cells, the lymphocyte subsets that pose major barriers to allogeneic and xenogeneic transplants. Current understanding of the mechanisms involved in tolerization of each cell type is reviewed. Considerable advances have been made in reducing the potential toxicity of conditioning regimens required for the induction of mixed chimerism in rodent models, and translation of these strategies to large animal models and in a patient are important advances toward more widespread clinical application of the mixed chimerism approach for tolerance induction.
Collapse
Affiliation(s)
- M Sykes
- Transplantation Biology Research Center, Surgical Service, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA.
| | | |
Collapse
|
383
|
Waldmann H, Hale G, Cobbold S. Appropriate targets for monoclonal antibodies in the induction of transplantation tolerance. Philos Trans R Soc Lond B Biol Sci 2001; 356:659-63. [PMID: 11375069 PMCID: PMC1088453 DOI: 10.1098/rstb.2001.0841] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
There are many routes to exploiting tolerance processes to ensure long-term graft survival. Complete tolerance although attractive as a goal, may not be the most practical in the clinic. Instead simple and low-impact procedures that harness tolerance processes used in conjunction with low doses of immunosuppressive drugs may prove the most reliable and user-friendly of approaches.
Collapse
Affiliation(s)
- H Waldmann
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK.
| | | | | |
Collapse
|
384
|
Abstract
Basic research has provided substantial encouragement that tolerance processes may be harnessed to the benefit of organ transplants. The goal of achieving mixed chimerism to ensure a robust tolerance, however elegant, may yet prove to be too complex and, consequently, risky as a procedure to compensate for the breadth of genetic differences, and prior immunological experiences of donor and host. Tolerance through regulation may prove to be easier to generate, but insufficiently robust to maintain. In the end the chosen protocol will be one that is simple, cheap and can guarantee patient compliance. Pragmatically, such a protocol need not be one aimed at clear-cut drug-free tolerance, but rather one which is trouble free. This could end up as a combination of partial tolerance (where tolerance processes have been harnessed) induced through a short-term treatment, in conjunction with easily tolerated maintenance therapy with select immunosuppressive drugs. Perhaps to the patient, the holy grail of tolerance is not as important as the complete assurance that the graft will survive and continue to function in all circumstances.
Collapse
Affiliation(s)
- H Waldmann
- Sir William Dunn School of Pathology, Oxford, UK.
| | | |
Collapse
|
385
|
|
386
|
|
387
|
Abstract
Although short- and long-term results after organ transplantation have improved considerably in recent years, morbidity and mortality rates in graft recipients remain high. The induction of lifelong donor-specific tolerance would dramatically improve outcome after organ transplantation. Although many tolerance protocols have been successful in rodent studies, most of these approaches have failed when attempted in large animals or humans. Robust tolerance, in contrast, has been demonstrated with mixed chimerism regimens not only in rodents but also in large animal models, including non-human primates. Furthermore, mixed chimerism protocols have been developed that would be feasible in cadaveric, and thus in thoracic, transplantation. The induction of mixed hematopoietic chimerism is therefore an attractive experimental approach for development of clinical tolerance protocols. One of the obstacles to widespread clinical application of this concept is the remaining toxicity of the host conditioning. Recent advances, however, have led to substantially milder protocols that could become clinically acceptable in the foreseeable future. This article provides a short overview of the basic mechanisms by which immunologic tolerance may be induced, describes the concept of mixed chimerism as a promising approach for clinical tolerance induction, and reviews recent progress in developing clinically feasible mixed chimerism protocols.
Collapse
Affiliation(s)
- T Wekerle
- Division of Transplantation, Department of Surgery, Vienna General Hospital, University of Vienna, Vienna, Austria.
| |
Collapse
|
388
|
Abstract
The immune system is naturally unresponsive to 'self' antigens. Improved knowledge of mechanisms underlying self tolerance is giving rise to a new generation of immunosuppressive agents, that can exploit these mechanisms and so reduce the nature and level of medication that needs to be given long-term to control diseases where the immune system does harm.
Collapse
Affiliation(s)
- H Waldmann
- Sir William Dunn School of Pathology, Oxford, UK.
| | | | | | | |
Collapse
|
389
|
Abstract
Immunotherapy of cancer with alloreactive lymphocytes is the mainstay of treatment, especially in haematological malignant disease. With donor lymphocyte infusion for immunotherapy, it is essential to induce host-versus-graft tolerance to ensure that the donor lymphocytes are accepted. Engraftment of haemopoietic cells of donor origin can be accomplished with reduced-intensity conditioning. Reducing transplant-related mortality by simplifing the stem-cell transplant procedure with a reduced-intensity regimen, particularly non-myeloablative conditioning, may have great potential for the treatment of malignant and non-malignant disorders.
Collapse
Affiliation(s)
- S Slavin
- Department of Bone Marrow Transplantation & Cancer Immunotherapy, Hadassah University Hospital, Ein Karem Jerusalem, Israel.
| |
Collapse
|
390
|
Adams AB, Durham MM, Kean L, Shirasugi N, Ha J, Williams MA, Rees PA, Cheung MC, Mittelstaedt S, Bingaman AW, Archer DR, Pearson TC, Waller EK, Larsen CP. Costimulation blockade, busulfan, and bone marrow promote titratable macrochimerism, induce transplantation tolerance, and correct genetic hemoglobinopathies with minimal myelosuppression. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:1103-11. [PMID: 11441122 DOI: 10.4049/jimmunol.167.2.1103] [Citation(s) in RCA: 138] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Mixed hemopoietic chimerism has the potential to correct genetic hemological diseases (sickle cell anemia, thalassemia) and eliminate chronic immunosuppressive therapy following organ transplantation. To date, most strategies require either recipient conditioning (gamma-irradiation, depletion of the peripheral immune system) or administration of "mega" doses of bone marrow to facilitate reliable engraftment. Although encouraging, many issues remain that may restrict or prevent clinical application of such strategies. We describe an alternative, nonirradiation based strategy using a single dose of busulfan, costimulation blockade, and T cell-depleted donor bone marrow, which promotes titratable macrochimerism and a reshaping of the T cell repertoire. Chimeras exhibit robust donor-specific tolerance, evidenced by acceptance of fully allogeneic skin grafts and failure to generate donor-specific proliferative responses in an in vivo graft-versus-host disease model of alloreactivity. In this model, donor cell infusion and costimulation blockade without busulfan were insufficient for tolerance induction as donor-specific IFN-gamma-producing T cells re-emerged and skin grafts were rejected at approximately 100 days. When applied to a murine beta-thalassemia model, this approach allows for the normalization of hemologic parameters and replacement of the diseased red cell compartment. Such a protocol may allow for clinical application of mixed chimerism strategies in patients with end-stage organ disease or hemoglobinopathies.
Collapse
MESH Headings
- Animals
- Antibodies, Blocking/administration & dosage
- B7-1 Antigen/immunology
- Bone Marrow Transplantation/immunology
- Busulfan/administration & dosage
- CD28 Antigens
- CD4-Positive T-Lymphocytes
- CD40 Antigens/immunology
- CD40 Ligand/immunology
- Cell Line
- Clonal Deletion/drug effects
- Clonal Deletion/genetics
- Cytotoxicity, Immunologic/drug effects
- Cytotoxicity, Immunologic/genetics
- Hemoglobinopathies/genetics
- Hemoglobinopathies/immunology
- Hemoglobinopathies/therapy
- Immunosuppression Therapy/adverse effects
- Injections, Intraperitoneal
- Lymphocyte Activation/immunology
- Male
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C3H
- Mice, Inbred C57BL
- Mice, SCID
- Radiation Chimera/immunology
- T-Lymphocytes, Cytotoxic/drug effects
- T-Lymphocytes, Cytotoxic/immunology
- Titrimetry
- Transplantation Tolerance/drug effects
- Transplantation Tolerance/genetics
- Transplantation Tolerance/immunology
Collapse
Affiliation(s)
- A B Adams
- Department of Surgery, The Carlos and Marguerite Mason Transplantation Biology Research Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
391
|
Taylor PA, Lees CJ, Waldmann H, Noelle RJ, Blazar BR. Requirements for the promotion of allogeneic engraftment by anti-CD154 (anti-CD40L) monoclonal antibody under nonmyeloablative conditions. Blood 2001; 98:467-74. [PMID: 11435318 DOI: 10.1182/blood.v98.2.467] [Citation(s) in RCA: 87] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The promotion of alloengraftment in the absence of global immune suppression and multiorgan toxicity is a major goal of transplantation. It is demonstrated that the infusion of a single modest bone marrow dosage in 200 cGy-irradiated recipients treated with anti-CD154 (anti-CD40L) monoclonal antibody (mAb) resulted in chimerism levels of 48%. Reducing irradiation to 100 or 50 cGy permitted 24% and 10% chimerism, respectively. In contrast, pan-T-cell depletion resulted in only transient engraftment in 200 cGy-irradiated recipients. Host CD4(+) cells were essential for alloengraftment as depletion of CD4(+) cells abrogated engraftment in anti-CD154-treated recipients. Strikingly, the depletion of CD8(+) cells did not further enhance engraftment in anti-CD154 mAb-treated recipients in a model in which rejection is mediated by both CD4(+) and CD8(+) T cells. However, anti-CD154 mAb did facilitate engraftment in a model in which only CD8(+) T cells mediate rejection. Furthermore, CD154 deletional mice irradiated with 200 cGy irradiation were not tolerant of grafts, suggesting that engraftment promotion by anti-CD154 mAb may not simply be the result of CD154:CD40 blockade. Together, these data suggest that a CD4(+) regulatory T cell may be induced by anti-CD154 mAb. In contrast to anti-CD154 mAb, anti-B7 mAb did not promote donor engraftment. Additionally, the administration of either anti-CD28 mAb or anti-CD152 (anti-CTLA-4) mAb or the use of CD28 deletional recipients abrogated engraftment in anti-CD154 mAb-treated mice, suggesting that balanced CD28/CD152:B7 interactions are required for the engraftment-promoting capacity of anti-CD154 mAb. These data have important ramifications for the design of clinical nonmyeloablative regimens based on anti-CD154 mAb administration.
Collapse
Affiliation(s)
- P A Taylor
- Department of Pediatrics, Division of Bone Marrow Transplantation, University of Minnesota Cancer Center, Minneapolis, USA
| | | | | | | | | |
Collapse
|
392
|
Bittencourt MC, Perruche S, Contassot E, Fresnay S, Baron MH, Angonin R, Aubin F, Hervé P, Tiberghien P, Saas P. Intravenous injection of apoptotic leukocytes enhances bone marrow engraftment across major histocompatibility barriers. Blood 2001; 98:224-30. [PMID: 11418484 DOI: 10.1182/blood.v98.1.224] [Citation(s) in RCA: 121] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cross-tolerization of T lymphocytes after apoptotic cell uptake by dendritic cells may be involved in self-tolerance maintenance. Furthermore, immunosuppressive properties are attributed to apoptotic cells. This study evaluated the consequences of apoptotic leukocyte administration in a restrictive engraftment model of murine bone marrow (BM) transplantation. Sublethally irradiated recipients received a limited number of allogeneic BM, with or without irradiated apoptotic leukocytes of different origins. No graft-versus-host disease was observed. Whereas only a low proportion of mice receiving BM cells alone engrafted, addition of apoptotic irradiated leukocytes, independently of the origin (donor, recipient, third-party mice, as well as xenogeneic peripheral blood mononuclear cells), significantly enhanced engraftment. Similar results were obtained after infusion of leukocytes rendered apoptotic by UVB irradiation or by anti-Fas monoclonal antibody stimulation, thus confirming the role of apoptotic cells in engraftment facilitation. Overall, these results suggest that apoptotic leukocytes can nonspecifically facilitate allogeneic BM engraftment. Such a simple approach could be of interest in BM transplantation settings involving an important HLA donor/recipient disparity, a T-cell-depleted graft, or reduced conditioning regimen intensity.
Collapse
Affiliation(s)
- M C Bittencourt
- Etablissement Français du Sang Bourgogne Franche-Comté, UPRES EA2284-Université de Franche-Comté, INSERM EO119 Besançon, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
393
|
Jin T, Toki J, Inaba M, Sugiura K, Fan T, Yu C, Lian Z, Takase K, Feng B, Ito T, Cui Y, Yang G, Ikehara S. A novel strategy for organ allografts using sublethal (7 Gy) irradiation followed by injection of donor bone marrow cells via portal vein. Transplantation 2001; 71:1725-31. [PMID: 11455249 DOI: 10.1097/00007890-200106270-00004] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
A new strategy for organ allografts that does not require recourse to immunosuppressants is established in mice. The strategy includes sublethal (7 Gy) irradiation followed by the injection of donor bone marrow cells (BMCs) via the portal vein (P.V.) and organ allografts 1 day after irradiation. Irradiation doses (< or =7 Gy) are found to allow the recipients to survive without the need to reconstitute the BMCs, as the recipient hematolymphoid cells can gradually recover. One hundred percent of recipients irradiated with 7 Gy followed by either P.V. or i.v. injection of donor BMCs accept organ allografts (the skin, pancreas, and adrenal glands) for more than 1 year. However, organ allograft survival rates decrease when irradiation doses are reduced; the skin graft survival rate of mice treated with 6.5 Gy and P.V. injection of BMCs is 79%, whereas that of mice treated with 6.5 Gy and i.v. injection is 50%, indicating that the P.V. injection of BMCs induces persistent tolerance more effectively than the i.v. injection. H-2 typing reveals that almost all the hematolymphoid cells (>98%) in the peripheral blood and hematolymphoid organs are donor-derived even 1 year after the treatment (7 Gy and P.V.). The T cells are tolerant to both donor-type and host-type MHC determinants. The major mechanism underlying the persistent tolerance induced by this strategy seems to be because of clonal deletion. This simple and safe strategy would be of great advantage for human organ transplantation.
Collapse
Affiliation(s)
- T Jin
- First Department of Pathology, Kansai Medical University, 10-15 Fumizono-cho, Moriguchi City, Osaka 570-8506, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
394
|
Abstract
Immunosuppressive drugs developed in the past two decades have improved the short-term survival of organ allografts, but tolerance has not been achieved and almost all transplant recipients continue to require drugs throughout life. Graft rejection arises from the cognate interaction of T cells with antigen-presenting cells, the recognition of alloantigen through the T-cell receptor, and the delivery of accessory stimulation signals. Once activated by the specific antigen, replicating T cells die if they are re-exposed to the same antigen. Since depletion of antigen-activated T cells is one critical mechanism of transplantation tolerance, drugs such as ciclosporin that interfere with activation-induced T-cell death could inhibit tolerance, whereas drugs such as mycophenolate mofetil, that induce the death of activated T cells, could facilitate tolerance. Other tolerance mechanisms depend on inactivation rather than elimination of allograft reactive T cells. When antigen recognition occurs without costimulation through the CD28 and CD154 accessory receptors, or in absence of cell division, T cells become unresponsive. Thus, inhibitors of CD28 and CD154, and inhibition of T-cell division by rapamycin promotes transplantation tolerance.
Collapse
Affiliation(s)
- X Yu
- Human Immunogenetics Program, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | | | | |
Collapse
|
395
|
Alwayn IP, Buhler L, Appel JZ, Goepfert C, Csizmadia E, Correa L, Harper D, Kitamura H, Down J, Awwad M, Sackstein R, Cooper DK, Robson SC. Mechanisms of thrombotic microangiopathy following xenogeneic hematopoietic progenitor cell transplantation. Transplantation 2001; 71:1601-9. [PMID: 11435972 DOI: 10.1097/00007890-200106150-00020] [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: 11/26/2022]
Abstract
INTRODUCTION Attempts to induce tolerance though mixed hematopoietic chimerism in the discordant pig-to-baboon xenotransplantation model are sometimes complicated by a potentially fatal thrombotic microangiopathy in the recipient baboons. This state develops immediately after the infusion of porcine mobilized peripheral blood leukocytes, containing progenitor cells (PBPC). In our study, we examined the interaction of infused porcine PBPC with recipient platelets in vivo in baboons and investigated the underlying mechanisms using an in vitro model. METHODS Two naïve baboons and six baboons preconditioned with irradiation and immunosuppression that received porcine PBPC were evaluated in vivo. The interaction of porcine and baboon PBPC with baboon platelets was investigated by an in vitro platelet aggregation assay. Fresh and cryopreserved PBPC were evaluated as well as PBPC obtained from growth-factor mobilized and unmobilized pigs. Furthermore, cellular subsets of PBPC were assessed for potential to induce platelet aggregation. Immunohistochemical staining was performed on platelet-leukocyte aggregates and potential inhibition of aggregation with anti-P-selectin and anti-CD154 mAbs, or eptifibatide (a GPIIb/IIIa receptor antagonist), was tested. RESULTS All baboons that received porcine PBPC rapidly developed marked thrombocytopenia (<20,000/microl), elevated serum lactate dehydrogenase (>1,500U/liter), schistocytosis, and platelet aggregates on blood smear. Three baboons died (two untreated and one preconditioned), and substantive platelet aggregates containing porcine leukocytes were observed in the microvasculature of lungs and kidneys. In vitro, porcine, but not baboon, PBPC induced aggregation of baboon platelets in a dose-dependent manner. Immunohistological examination of these aggregates confirmed the incorporation of porcine leukocytes. Cryopreserved PBPC caused less aggregation than fresh PBPC, and growth-factor-mobilized PBPC induced less aggregation than unmobilized PBPC. Aggregation was fully abrogated by the addition of eptifibatide, and modulated by anti-P-selectin and anti-CD154 monoclonal antibodies that recognize adhesion receptors on activated platelets. Purified fractions (granulocytes, CD2+, and CD- cells) of porcine PBPC did not initiate aggregation, whereas addition of exogenous porcine PBPC membranes (erythrocytes, dead cells, and/or platelets) to the purified fractions exacerbated the aggregation response. CONCLUSIONS These data indicate that porcine PBPC mediate aggregation of baboon platelets. This process likely contributes to the thrombotic microangiopathy observed after PBPC transplantation in the pig-to-baboon model. Eptifibatide can fully abrogate platelet aggregation induced by porcine PBPC in vitro. Purification of the progenitor cells from porcine PBPC and/or treatment of baboons with eptifibatide may be beneficial.
Collapse
Affiliation(s)
- I P Alwayn
- The Transplantation Biology Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
396
|
Taylor PA, Noelle RJ, Blazar BR. CD4(+)CD25(+) immune regulatory cells are required for induction of tolerance to alloantigen via costimulatory blockade. J Exp Med 2001; 193:1311-8. [PMID: 11390438 PMCID: PMC2193378 DOI: 10.1084/jem.193.11.1311] [Citation(s) in RCA: 476] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2000] [Accepted: 03/20/2001] [Indexed: 11/18/2022] Open
Abstract
Immune regulatory CD4(+)CD25(+) cells play a vital role in the induction and maintenance of self-tolerance and are essential for T cell homeostasis and the prevention of autoimmunity. Induction of tolerance to allogeneic donor grafts is a clinically desirable goal in bone marrow and solid organ transplantation. To determine whether CD4(+)CD25(+) cells regulate T cell responses to alloantigen and are critical for tolerance induction, murine CD4(+) T cells were tolerized to alloantigen via ex vivo CD40 ligand (CD40L)/CD40 or CD28/cytotoxic T lymphocyte-associated antigen 4/B7 blockade resulting in secondary mixed leukocyte reaction hyporesponsiveness and tolerance to alloantigen in vivo. CD4(+)CD25(+) T cells were found to be potent regulators of alloresponses. Depletion of CD4(+)CD25(+) T cells from the CD4(+) responder population completely abrogated ex vivo tolerance induction to alloantigen as measured by intact responses to alloantigen restimulation in vitro and in vivo. Addback of CD4(+)CD25(+) T cells to CD4(+)CD25(-) cultures restored tolerance induction. These data are the first to indicate that CD4(+)CD25(+) cells are essential for the induction of tolerance to alloantigen and have important implications for tolerance-inducing strategies targeted at T cell costimulatory pathways.
Collapse
Affiliation(s)
- Patricia A. Taylor
- University of Minnesota Cancer Center and Department of Pediatrics, Division of Bone Marrow Transplantation, Minneapolis, Minnesota 55455
| | - Randolph J. Noelle
- Department of Microbiology, Dartmouth Medical College, Hanover, New Hampshire 03756
| | - Bruce R. Blazar
- University of Minnesota Cancer Center and Department of Pediatrics, Division of Bone Marrow Transplantation, Minneapolis, Minnesota 55455
| |
Collapse
|
397
|
Abstract
Achieving transplantation tolerance is an important goal in the effort to reduce long-term morbidity and mortality in organ transplant recipients. Robust, lifelong, donor-specific tolerance can be reliably achieved by induction of mixed chimerism in various animal models. To date, the clinical application of these proto-cols has been impeded partly by the potential toxicity of the required host conditioning regimens and the lack of successful studies in large animals. This article reviews the progress achieved in recent years in developing considerably milder conditioning protocols in rodents, and in extending some of these models to achieve permanent mixed chimerism and tolerance in large animals. Advances in the induction of xenogeneic tolerance through mixed chimerism are also discussed.
Collapse
Affiliation(s)
- T Wekerle
- Department of Surgery, Vienna General Hospital, University of Vienna, Währingergürtel 18, Vienna, A-1090 Austria.
| | | |
Collapse
|
398
|
Auchincloss H. In search of the elusive Holy Grail: the mechanisms and prospects for achieving clinical transplantation tolerance. Am J Transplant 2001; 1:6-12. [PMID: 12095040 DOI: 10.1034/j.1600-6143.2001.010103.x] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- H Auchincloss
- Transplantation Unit, Massachusetts General Hospital, Boston 02114, USA.
| |
Collapse
|
399
|
Dantal J, Josien R, Soulillou JP. Advances in transplant immunobiology. Curr Opin Nephrol Hypertens 2001; 10:349-54. [PMID: 11342796 DOI: 10.1097/00041552-200105000-00008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The present review briefly addresses the most recent knowledge acquired in the field of transplant immunology. A particular emphasis is placed on articles published during the past 12-18 months that have focused on allorecognition, dendritic cells and tolerance.
Collapse
Affiliation(s)
- J Dantal
- Institut de Transplantation et de Recherche en Transplantation, CHR Nantes Hotel Dieu, Nantes, France
| | | | | |
Collapse
|
400
|
Li S, Thanikachalam M, Pang M, Carreno M, Aitouche A, Pham SM. Combined host-conditioning with CTLA4-Ig, tacrolimus, anti-lymphocyte serum, and low-dose radiation leads to stable mixed hematopoietic chimerism. Exp Hematol 2001; 29:534-41. [PMID: 11301194 DOI: 10.1016/s0301-472x(00)00685-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The toxic dose of irradiation required to achieve stable mixed hematopoietic chimerism is the major limitation to its clinical application in transplantation and other nonmalignant conditions such as hemoglobinopathies. This study examines the additive effect of costimulatory blockage, to our previously described tacrolimus-based conditioning regimen, in further reducing the dose of total-body irradiation to achieve stable mixed chimerism in rats. Fully mismatched, 4- to 6-week-old ACI and Wistar Furth rats were used as donors and recipients, respectively. Recipients were administered CTLA4-Ig 2mg/kg/day (alternate days) in combination with tacrolimus 1 mg/kg/day (daily) from day 0 through day +10, anti-lymphocyte serum 10 mg at day +10 (single dose), and total-body irradiation ranging from 100-600 cGy, prior to bone marrow transplantation (day 0) with 100 x 10(6) of T-cell-depleted bone marrow cells. Levels of donor chimerism were determined over a period of 12 months. The short course of CTLA4-Ig, tacrolimus, and ALS led to dramatic engraftments at reduced doses of irradiation: 100% (5/5) and 93% (13/14) of the animals developed mixed chimerism at 400 cGy and 300 cGy, respectively. At 300 cGy, recipients exhibited durable, multilineage mixed chimerism at 365 days with donor cells ranging from 19-42% (mean 23.4%) with no evidence of graft-vs-host disease. These mixed chimeras exhibited in vitro (mixed lymphocyte reaction) and in vivo (skin grafts) donor-specific tolerance. This study suggests that addition of costimulatory blockade to a tacrolimus-based conditioning regimen reduces the dose of irradiation required to achieve stable multilineage chimerism in rats.
Collapse
Affiliation(s)
- S Li
- Department of Surgery, University of Miami School of Medicine, 1801 N.W. 9th Ave., Miami, FL 33136, USA
| | | | | | | | | | | |
Collapse
|