201
|
Filipec M, Hasková Z, Havrlíková K, Letko E, Holán V, Matousek J, Kalousek I. Immunosuppressive effect of bovine seminal ribonuclease on a model of corneal transplantation in rabbit. Graefes Arch Clin Exp Ophthalmol 1996; 234:586-90. [PMID: 8880158 DOI: 10.1007/bf00448804] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Bovine seminal ribonuclease (BS RNase) was determined to have a specific suppressive effect on the proliferation of T lymphocytes in vitro. Its immunosuppressive effect was proven in skin grafting in mice as well. METHODS The immunosuppressive effect of BS RNase was evaluated in tissue cultures and on a model of corneal transplantation in rabbits. The penetration of BS RNase into the anterior chamber was detected by immunoblotting of anterior chamber fluid obtained from animals treated either topically or subconjunctivally. RESULTS In vitro blastic transformation of mouse T lymphocytes was significantly inhibited by BS RNase (concentrations 15-250 micrograms/ml). No such effect was observed on B lymphocytes. In the rabbit model of corneal graft rejection, BS RNase injected subconjunctivally prolonged mean graft survival time significantly (33.4 days) compared with placebo (salt solution; MST 17.7 days). No BS RNase was detected by immunoblotting in anterior chamber fluid after either topical or subconjunctival application. CONCLUSION BS RNase showed significant immunosuppressive effect both in the blastic transformation test and in the rabbit high-risk model of corneal transplantation. Negative results of anterior chamber fluid immunoblotting indicate poor absorption of the drug.
Collapse
Affiliation(s)
- M Filipec
- Second Department of Ophthalmology, First Medical Faculty, Charles University, Prague, Czech Republic
| | | | | | | | | | | | | |
Collapse
|
202
|
Clement JD, Chan SY, Bishop DK. Allogeneic class I MHC requirement for alloantigen-reactive helper T-lymphocyte responses in vivo. Evidence for indirect presentation of alloantigen. Transplantation 1996; 62:388-96. [PMID: 8779688 DOI: 10.1097/00007890-199608150-00015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The mechanisms by which host T cells recognize transplant-associated alloantigens in vivo have not been established. Two alloantigen presentation pathways may be used: (1) allogeneic class I and class II MHC molecules may be recognized directly by host CD8+ and CD4+ cells, respectively, or (2) allogeneic MHC molecules may be processed as foreign peptide and presented by host antigen-presenting cells to CD4+ cells in the context of self class II proteins. In this study, the sponge matrix allograft model was used to examine the relative contributions of these alloantigen presentation pathways to CD4+ T-cell activation in vivo. Limiting dilution analysis was used to quantify the localization of interleukin-2-producing helper T lymphocytes (HTL) following implantation of sponge allografts. Allografts either were disparate at both class I and class 11, or were derived from beta2-microglobulin knockout (beta2M-/-) mice, which express class II but are deficient in class I. Two measures of in vivo HTL function were monitored: (1) the accumulation of HTL within the allograft (a process that is dependent upon antigen-driven cytokine production), and (2) the development of cytolytic alloantibodies. After implantation of sponge allografts expressing both class I and class II, HTL were readily detectable in the allograft, and cytolytic alloantibodies were present in the serum. When mice were implanted with beta2M-/- sponge allografts, HTL failed to infiltrate these class I-deficient allografts, and alloantibodies were not detectable in the sera of recipients of beta2M-/- sponge allografts. This in vivo requirement for class I expression was not reflected by traditional in vitro measures of HTL function; cells obtained from lymphoid tissues mounted a mixed lymphocyte response and produced interleukin-2 when stimulated with beta2M-/- splenocytes in vitro. One possible interpretation of these data is that in vivo HTL functions are dependent upon the presence of class I-reactive CD8+ T cells. However, HTL readily infiltrated grafts expressing both class I and class II when recipients depleted of CD8+ T cells, and alloantibodies were produced. These observations support the idea that indirect presentation of allogeneic class I molecules plays a critical role in regulating CD4+ HTL functions associated with allograft rejection in vivo.
Collapse
Affiliation(s)
- J D Clement
- Department of Internal Medicine, University of Michigan, Ann Arbor 48109, USA
| | | | | |
Collapse
|
203
|
Abstract
The basic immunosuppressive protocol used in most transplant centers involves the use of multiple drugs, each directed at a discrete site in the T-cell activation cascade and each with distinct side effects. Cyclosporine, azathioprine, corticosteroids, FK506 (tacrolimus), and RS61443 (mycophenolate mofetil) have been approved by the Food and Drug Administration, and the clinical efficacy of rapamycin (sirolimus), mizoribine, 15-deoxyspergualin, and leflunomide is being explored. Based on their primary site of action, the immunosuppressants can be classified as inhibitors of transcription (cyclosporine, tacrolimus), inhibitors of nucleotide synthesis (azathioprine, mycophenolate mofetil, mizoribine, leflunomide), inhibitors of growth factor signal transduction (sirolimus, leflunomide), and inhibitors of differentiation (15-deoxyspergualin). Polyclonal antilymphocyte antibodies, monoclonal antibodies directed at the T-cell antigen receptor complex (OKT3, TIOB9), and monoclonal antibodies directed at additional cell surface antigens, including interleukin-2 receptor alpha, afford cell-specific regulation of the immune response and are being used in the clinical setting as induction therapy and/or antirejection drugs. Clearly, the transplant clinician now has a greater choice in the selection and application of immunosuppressants in the clinic for the fine regulation of the antiallograft repertory. The prevailing paradigm regarding the mechanisms of action of immunosuppressants is that they all function to prevent allograft rejection by preventing/inhibiting cell activation, cytokine production, differentiation, and/or proliferation. One hypothesis, albeit provocative, is that some of the immunosuppressants might function by stimulating the expression of immunosuppressive molecules and/or cells.
Collapse
Affiliation(s)
- M Suthanthiran
- Rogosin Institute, Department of Transplantation Medicine and Extracorporeal Therapy, New York Hospital-Cornell Medical Center, New York, USA
| | | | | |
Collapse
|
204
|
|
205
|
Murphy B, Sayegh MH. Why do we reject a graft? Mechanisms of recognition of transplantation antigens. Transplant Rev (Orlando) 1996. [DOI: 10.1016/s0955-470x(96)80024-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
206
|
Roelen DL, Dover EL, Niimi M, Young NT, Morris PJ, Wood KJ. Semi-allogeneic (F1) versus fully allogeneic blood transfusions: differences in their ability to induce specific immunological unresponsiveness. Eur J Immunol 1996; 26:1468-74. [PMID: 8766548 DOI: 10.1002/eji.1830260710] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The beneficial effect on graft survival achieved by pretransplant blood transfusions is well established. However, the type of major histocompatibility complex (MHC) mismatch between transfusion donor and recipient seems to play a role in determining the outcome. The hypothesis that this sharing of MHC antigens is correlated with the level of sensitization or tolerization was studied in mice by pretreatment with semi-allogeneic (F1) or with fully allogeneic whole blood transfusions. Limiting dilution analysis (LDA) in vitro for donor-specific T helper (Thp) and cytotoxic T lymphocyte precursors (CTLp) performed on splenocytes isolated from transfused recipients 2 or 4 weeks after transfusion showed that the duration and magnitude of the response was reduced after a semi-allogeneic compared to a fully allogeneic transfusion. After a semi-allogeneic transfusion, both Thp and CTLp frequencies had returned to naive levels 4 weeks after transfusion, whereas after infusion of fully allogeneic blood, they remained elevated after 4 weeks. When a fully allogeneic heart was transplanted 2 or 4 weeks after transfusion, a small but significant improvement in graft prolongation (2 weeks, not significant, 4 weeks: p < 0.01) was observed following pretreatment with a semi-allogeneic transfusion (2 weeks: median survival time (MST) 30 days, 4 weeks: MST 29 days) compared to that obtained after fully allogeneic transfusion (2 weeks: MST 23 days, 4 weeks: MST 12 days). The semi-allogeneic transfusions were correlated with a statistically significant prolonged (7 days) persistence of donor-derived MHC class II+ cells in the recipient and with reduced levels of anti-donor MHC class I-specific antibody formation compared to these responses after transfusion with fully allogeneic cells. These results demonstrate that pretreatment with a semi-allogeneic blood transfusion is more tolerizing and less sensitizing than pretreatment with a fully allogeneic blood transfusion. These findings may be explained by the sharing of MHC antigens between recipient and transfusion donor.
Collapse
Affiliation(s)
- D L Roelen
- University of Oxford, Nuffield Department of Surgery, John Radcliffe Hospital, Headington, GB.
| | | | | | | | | | | |
Collapse
|
207
|
Duquesnoy RJ, Liu K, Moliterno RA, Attfield D. Do heat shock proteins play a role in transplant immunity? Transplant Rev (Orlando) 1996. [DOI: 10.1016/s0955-470x(96)80026-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
208
|
Duncan SR, Valentine V, Roglic M, Elias DJ, Pekny KW, Theodore J, Kono DH, Theofilopoulos AN. T cell receptor biases and clonal proliferations among lung transplant recipients with obliterative bronchiolitis. J Clin Invest 1996; 97:2642-50. [PMID: 8647959 PMCID: PMC507352 DOI: 10.1172/jci118714] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Obliterative bronchiolitis (OB) is the most serious late complication of lung transplantation, but the pathogenesis of this disorder has not been elucidated. We sought evidence that OB is mediated by a cellular immunologic response by characterizing T cell antigen receptor beta-chain variable gene (TCRBV) repertoires in lung allograft recipients. Expression levels of 27 TCRBV among recipients were determined by multiprobe RNase protection assay after PCR amplification. In comparison to recipients with no evidence of rejection (n = 9), the PBL TCRBV repertoires of OB subjects (n = 16) exhibited more frequent expansions (16 vs. 9% of all measured TCRBV, P < 0.02), and the magnitudes of these abnormalities were greater (8.2 +/- 0.8 vs. 4.5 +/- 0.3 SD from mean normal values, P < 0.01). TCRBV sequencing showed these expansions were composed of clonal or oligoclonal populations. Thus, T cell responses in the recipients are marked by highly selective clonal expansions, presumably driven by indirect recognition of a limited number of immunodominant alloantigens. These processes are exaggerated among allograft recipients with OB, implying that cognate immune mechanisms are important in the pathogenesis of the disorder. Furthermore, the prominence of finite, distinct TCR phenotypes raise possibilities for development of novel diagnostic modalities and targeted immunotherapies for OB and other manifestations of chronic allograft rejection.
Collapse
Affiliation(s)
- S R Duncan
- Department of Immunology, Scripps Research Institue, La Jolla, California 92037, USA
| | | | | | | | | | | | | | | |
Collapse
|
209
|
|
210
|
Shoskes DA, Halloran PF. Delayed graft function in renal transplantation: etiology, management and long-term significance. J Urol 1996; 155:1831-40. [PMID: 8618268 DOI: 10.1016/s0022-5347(01)66023-3] [Citation(s) in RCA: 227] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
PURPOSE In cadaveric renal transplantation a period of delayed graft function postoperatively is not uncommon and often associated with a poor outcome. We reviewed the biology of reperfusion injury and delayed graft function in renal transplantation, as well as its prevention, management and long-term effects. MATERIALS AND METHODS The medical literature covering acute tubular necrosis, delayed graft function in renal transplantation and immunology of ischemia reperfusion injury was reviewed. RESULTS Delayed graft function is clearly associated with poor allograft survival, and is caused by an interaction of ischemic and immunological factors. Technical and pharmacological maneuvers can improve early function rates. The response to ischemic injury is complex, and may increase graft immunogenicity and promote the chronic proliferative changes seen in chronic allograft nephropathy. CONCLUSIONS Improvement in early renal function should be a primary goal in renal transplantation to enhance early and long-term results. Basic research into the injury response may yield insights into renal pathophysiology.
Collapse
Affiliation(s)
- D A Shoskes
- Department of Surgery, UCLA School of Medicine, Harbor-UCLA Medical Center, Torrance, USA
| | | |
Collapse
|
211
|
Ostraat O, Riesbeck K, Qi Z, Eriksson T, Schatz H, Ekberg H. Thalidomide prolonged graft survival in a rat cardiac transplant model but had no inhibitory effect on lymphocyte function in vitro. Transpl Immunol 1996; 4:117-25. [PMID: 8843588 DOI: 10.1016/s0966-3274(96)80005-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The effects of thalidomide on in vitro interleukin 2 (IL-2) production and thymidine uptake by human peripheral blood lymphocytes or rat splenocytes were investigated. Phytohaemagglutinin-stimulated human lymphocytes were incubated in the presence of thalidomide added at culture initiation. No immunosuppressive effect of thalidomide was observed in these experiments. Primary human mixed lymphocyte cultures treated with thalidomide for 6 days were also unaffected. A microsomal rabbit liver homogenate was prepared for metabolizing thalidomide. Stimulated lymphocytes secreted significantly more IL-2 in the presence of microsomal-treated thalidomide than did controls. The effect of thalidomide was then studied either as single therapy or in combination with cyclosporin A (CyA) in a rat allograft cardiac transplantation model. In addition, T cell subsets were analysed by flow cytometry in untransplanted rats treated with thalidomide. Treatment was given as induction therapy from the day of transplantation until day 9. Graft survival in rats treated with thalidomide was significantly prolonged compared to the untreated group. No difference in graft survival was detected between rats treated with thalidomide or CyA only. Graft survival was found to be slightly prolonged in rats given thalidomide and CyA in combination compared to rats treated with CyA alone. In untransplanted rats given thalidomide a decrease of CD4 positive T cells was detected on days 3 and 5. The T helper/cytotoxic-suppressor cell ratio was significantly diminished but, after 1 week of treatment, values for T cell subsets had almost returned to baseline levels. No inhibitory effect was obtained when phytohaemagglutinin-stimulated rat splenocytes were cultured with metabolized thalidomide. In summary, the ability of thalidomide to improve allograft survival in a solid organ transplant model was verified. The occurrence of thalidomide-induced changes in T cell subset ratios was demonstrated. In in vitro studies, however, there was no decrease but an increase in IL-2 production, and no change in thymidine uptake. The mechanism responsible for the immunosuppressive effect of thalidomide remains to be elucidated.
Collapse
Affiliation(s)
- O Ostraat
- Department of Surgery, University Hospital, Malmö, Sweden
| | | | | | | | | | | |
Collapse
|
212
|
Dorling A, Lombardi G, Binns R, Lechler RI. Detection of primary direct and indirect human anti-porcine T cell responses using a porcine dendritic cell population. Eur J Immunol 1996; 26:1378-87. [PMID: 8647220 DOI: 10.1002/eji.1830260630] [Citation(s) in RCA: 98] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The pathways of human anti-pig T cell xenorecognition have been investigated. Freshly isolated porcine alveolar lavage (AL) cells induced primary proliferative responses by human peripheral and cord blood mononuclear cells which were inhibited by anti-HLA-DR antibody (indirect xenorecognition). Following over-night culture, the AL cells acquired the capacity to stimulate proliferation by purified human T cells which was inhibited by anti-SLA-DR antibody (direct xenorecognition). The marked increase in immunogenicity in the porcine AL cells was accompanied by a phenotypic change consistent with dendritic cell maturation. Limiting dilution assays indicate that the total anti-pig T cell response, in particular that mediated by indirect xenorecognition, is stronger than comparable alloresponses.
Collapse
Affiliation(s)
- A Dorling
- Department of Immunology, Royal Postgraduate Medical School, Hammersmith Hospital, London, GB
| | | | | | | |
Collapse
|
213
|
Saitovitch D, Morris PJ, Wood KJ. Recipient cells expressing single donor MHC locus products can substitute for donor-specific transfusion in the induction of transplantation tolerance when pretreatment is combined with anti-Cd4 monoclonal antibody. Evidence for a vital role of Cd4+ T cells in the induction of tolerance to class I molecules. Transplantation 1996; 61:1532-8. [PMID: 8633384 DOI: 10.1097/00007890-199605270-00021] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Syngeneic cells expressing single donor MHC locus products have been shown to induce specific immunological hyporesponsiveness, but not tolerance, to an allograft. In this study we have attempted to potentiate the beneficial effect of pretreatment with single donor MHC locus products by the addition of anti-Cd4(2) monoclonal antibody to the pretreatment protocol. We show that pretreatment with recipient L cells expressing the products of a single donor locus (K, D, or IA) can induce tolerance to a C57BL/10 (H2b) cardiac allograft in C3H/He (H2k) mice, when given in combination with the depleting anti-Cd4 monoclonal antibody YTA 3.1.2. Both the induction and maintenance phases of tolerance were found to be antigen-specific. Cells expressing donor class II antigens, IA(b) were found to be most effective. Interestingly, donor class I molecules were also found to be capable of inducing specific unresponsiveness in combination with anti-Cd4, provided an optimal antigenic load was delivered at the time of pretreatment. Pretreatment with cells expressing donor class I and anti-Cd8 monoclonal antibody had no beneficial effect on graft survival. These data show that single donor MHC locus products can induce immunologic tolerance to fully MHC and minor histocompatibility antigen-mismatched heart grafts when given under the cover of anti-Cd4 mAb. They also show that Cd4+ T cells play an important role in the induction of specific unresponsiveness to class I alloantigen in vivo and suggest that the blockade of T cells capable of recognizing class I alloantigen presented indirectly is important in the induction of tolerance.
Collapse
Affiliation(s)
- D Saitovitch
- Nuffield Department of Surgery, University of Oxford, John Radcliffe Hospital, Headington, Oxford, England
| | | | | |
Collapse
|
214
|
Abstract
The key to understanding afferent immunity is the mechanism of activation of T lymphocytes by specialized antigen presenting cells, which bind antigenic peptide to Class II major histocompatibility molecules, and stimulate T cells via Signal 1 (antigen) and Signal 2 (costimulation). The best studied costimulatory pathway is the interaction of B7-1 or B7-2 ligand molecules on antigen presenting cells with CD28 or CTLA-4 receptors on T cells. T cell signaling occurs through the T cell receptor-CD3 complex and is augmented by cosignaling via CD4, CD8, and CD45. The activation of T cells to alloantigen occurs by either a direct pathway of recognition of allogenic major histocompatibility molecules (with or without an associated endogenous peptide), or by an indirect pathway of recognition of processed donor alloantigens via recipient antigen presenting cells. Afferent immunity on the musculoskeletal system is of special interest because of the absence of viable donor antigen presenting cells in processed grafts that makes them susceptible to the indirect pathway of alloantigen recognition.
Collapse
Affiliation(s)
- A A Czitrom
- University of Texas, Southwestern Medical Center, Medical City Dallas Hospital, Advanced Surgical Institutes, USA
| |
Collapse
|
215
|
Murphy B, Auchincloss H, Carpenter CB, Sayegh MH. T cell recognition of xeno-MHC peptides during concordant xenograft rejection. Transplantation 1996; 61:1133-7. [PMID: 8610405 DOI: 10.1097/00007890-199604270-00001] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
T cell recognition of xenoantigens is likely to play a key role in rejection of xenografts surviving hyperacute and delayed xenograft rejection, but the mechanisms of how this might occur are unknown. We used synthetic rat class II MHC peptides to study the role of the indirect pathway, where processed xenogeneic MHC antigens are presented in the context of self MHC, in a concordant xenograft rejection model in vivo. Mice of four different strains, BALB/c, B1O.A, CBA/ca, and C57BL/6j, were immunized with a mixture of rat class II MHC 25-mer xenopeptides representing the full-length sequence of the beta chain hypervariable domain of either RT1.Du (DR and I-E like) or RT1.Bu (DQ and I-A like) of the Wistar-Furth (WF) (RT1u) rat. Draining lymph node cells were capable of recognizing and proliferating to specific class II xeno-MHC peptides. The immunogenicity of the different peptides varied with the responder mouse strain. Responder T cells were CD4+, and were inhibited by anti-I-A and anti-I-E antibodies. We then examined the proliferative response of T cells from B1O.A primed by WF skin or vascularized cardiac xenografts to the class II MHC xenopeptides, when presented by naive B1O.A splenic antigen-presenting cells. These T cells were capable of proliferating to the same xeno-MHC peptides shown to be immunogenic by immunization. These data confirm the occurrence of self-restricted T cell recognition of xeno-MHC peptides in xenograft rejection, and provide the rationale for further investigating the role of the indirect pathway of recognition in xenotransplantation.
Collapse
Affiliation(s)
- B Murphy
- Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
216
|
Fedoseyeva EV, Tam RC, Popov IA, Orr PL, Garovoy MR, Benichou G. Induction of T cell responses to a self-antigen following allotransplantation. Transplantation 1996; 61:679-83. [PMID: 8607166 DOI: 10.1097/00007890-199603150-00001] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
T cell tolerance to self-antigens is established through the recognition by immature T cells of dominant self-peptides presented in association with self-MHC molecules in the developing thymus (negative selection). The self-peptide Dd 61-80 is dominant in syngeneic BALB/c mice (H2d). T cell tolerance to Dd 61-80 in this mouse strain resulted in the absence of T cell proliferation following in vivo priming with Dd 61-80 peptide. Here, we show that transplantation of BALB/c mice with allogeneic B10.A (H2a) splenocytes led to an autoimmune T cell response toward the dominant self-peptide Dd 61-80. No T cell responses to Dd 61-80 peptide were observed after transplantation of C57BL/6 (H2b) splenocytes into BALB/c recipients. In addition, we provide evidence indicating that the breakdown of tolerance to Dd 61-80 self-peptide resulted from the presentation of the donor crossreactive peptide Kk 61-80 at the surface of recipient antigen-presenting cells. Taken together, our results suggest that following allotransplantation, T cell responses to donor antigens could spread to crossreactive determinants on self-proteins, thus perpetuating and amplifying the rejection process and presumably initiating tissue-specific autoimmune disorders.
Collapse
Affiliation(s)
- E V Fedoseyeva
- Immunogenetics and Transplantation Laboratory, Department of Surgery, University of California San Francisco, California 94143-0508 USA
| | | | | | | | | | | |
Collapse
|
217
|
Sano Y, Ksander BR, Streilein JW. Minor H, rather than MHC, alloantigens offer the greater barrier to successful orthotopic corneal transplantation in mice. Transpl Immunol 1996; 4:53-6. [PMID: 8762011 DOI: 10.1016/s0966-3274(96)80035-9] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Irrespective of HLA matching, a far higher proportion of human corneal allografts placed orthotopically in avascular corneal graft beds are accepted indefinitely, compared to other types of solid tissue allografts. However, many more corneal grafts are rejected if they are transplanted onto neovascularized recipient eyes. Using a murine model of orthotopic corneal transplantation in which grafts were placed in normal eyes, we have reported previously that grafts bearing minor H antigens alone are more likely to be rejected (approximately 50%) than are grafts displaying only MHC alloantigens (< 20%). Moreover, recipients of MHC plus minor H incompatible corneal grafts developed delayed hypersensitivity (DH) directed solely at minor H antigens. These studies have now been extended to include corneal grafts placed in neovascularized recipient eyes. Neovascularization was induced by placing sutures in the central cornea of one eye of BALB/c mice. Two weeks later corneas from C57BL/10 donors were grafted into these eyes. Rejection reactions were first apparent within 7 days and all grafts were destroyed by 14 days. Donor-specific DH responses were examined by injecting irradiated donor antigen-bearing spleen cells into the ear pinna. To distinguish DH directed at MHC versus minor antigens, some graft recipients were ear-challenged with BALB.B cells (donor MHC only), while other received B10.D2 cells (donor minor H only). Intense ear-swelling responses were evoked by B10.D2 cells, but not by BALB.B cells. These findings indicate that, for orthotopic corneal allografts, minor H antigens offer a more formidable barrier to graft acceptance than do MHC-encoded antigens. We speculate that this unexpected outcome may reflect a reduced level of MHC expression on corneal tissue. Moreover since the cornea lacks bone marrow derived dendritic cells, allorecognition by recipient T cells must occur via the indirect pathway, and in this situation minor H antigens may compete favorably with MHC antigens for processing and presentation by recipient antigen-presenting cells.
Collapse
Affiliation(s)
- Y Sano
- Schepens Eye Research Institute, Boston, MA 02114, USA
| | | | | |
Collapse
|
218
|
Sayegh MH, Khoury SJ, Hancock WW, Weiner HL, Carpenter CB. Mechanisms of oral tolerance by MHC peptides. Ann N Y Acad Sci 1996; 778:338-45. [PMID: 8610987 DOI: 10.1111/j.1749-6632.1996.tb21141.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Recent evidence indicates that MHC peptides play an important role in T-cell recognition of alloantigen. We studied the tolerogenicity of orally administered synthetic MHC allopeptides in the rat model. Initially, we demonstrated that oral administration of synthetic class II MHC allopeptides significantly inhibited the DTH response to the peptides as well as to donor-derived cells. The tolerogenic effect was antigen specific and was induced by immunogenic, but not by nonimmunogenic, allopeptides. Immunohistological studies of DTH skin lesions showed that oral tolerance is associated with a state of "immune deviation" to a predominance of Th2 cell function in the lesions. We recently extended the above observations and examined the tolerogenic effect of orally administered synthetic MHC allopeptides on MLR and CTL generation. We found that oral administration of the class II allopeptides effected significant reduction of MLR proliferation and CTL generation, which was antigen specific. In addition, similar to the DTH results when we compared the tolerogenicity of the immunogenic versus the nonimmunogenic peptides, MLR and CTL suppression was significantly higher with the immunogenic peptides. The gut immune system play an important role in oral tolerance by MHC peptides. Initial experiments showed that intestinal epithelial cells pulsed in vitro with immunogenic MHC allopeptides, or in vivo by oral administration of immunogenic peptides, were capable of presenting these peptides to primed T cells in vitro. Whether such presentation by intestinal epithelial cells or other gut antigen-presenting cells leads to preferential activation of Th2 regulatory cells, which ultimately suppress Th1 alloimmune responses, remains to be determined.
Collapse
Affiliation(s)
- M H Sayegh
- Laboratory of Immunogenetics and Transplantation, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | |
Collapse
|
219
|
SUTHANTHIRAN M, STROM TB. Human renal allograft rejection: Immune mechanisms, molecular correlates and treatment strategies. Nephrology (Carlton) 1996. [DOI: 10.1111/j.1440-1797.1996.tb00060.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
220
|
Gill RG, Coulombe M, Lafferty KJ. Pancreatic islet allograft immunity and tolerance: the two-signal hypothesis revisited. Immunol Rev 1996; 149:75-96. [PMID: 9005221 DOI: 10.1111/j.1600-065x.1996.tb00900.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The principle assumption of this discussion is that costimulation (CoS) forms the primary stimulus that compels T cells to mount a response to their specific antigen. However, this response can be either positive or negative, depending on the developmental stage of the T cell and the microenvironment in which the antigen and CoS are received. Thus, both immunity and tolerance may represent different outcomes of a two-signal process. We would emphasize that CoS is a functional term and not a strict molecular definition. While many molecular interactions have been described as providing CoS activity, notably those involving the B-7 family of cell surface molecules, it is not yet clear what combination(s) of non-antigen-specific signals may fulfil this function. This point is important because many studies have achieved tolerance through strategies designed to inhibit specific CoS molecules. However, it may be that differential signaling through distinct CoS molecules, rather than a global inhibition of CoS per se, plays a role in the generation of active tolerance in such studies (Bluestone 1995). A corollary of this notion is that antigen (signal 1) delivery to T cells is a null event and so is not an inherently paralysing signal. Of course, if signal 1 is not itself a tolerogenic signal, then other mechanisms are necessary to explain many empirical observations of tolerance to allogeneic or self antigens. This is best illustrated by those forms of functional tolerance to either alloantigens or self antigens that do not appear to be the result of clonal deletion/inactivation. It would be relatively simple to invoke a model of tolerance whereby the relevant tissue-destructive cell is eliminated or inactivated; such a model would preclude the necessity to suggest active regulatory mechanisms of tolerance. However, in several model systems, including our own observations concerning tolerance induction to APC-depleted islet allografts, tissue-destructive T cells can persist in recipients tolerant to allogeneic or self antigens. Furthermore, there are key examples in which tolerance demonstrates a dominant phenotype; that is, tolerant cells can regulate the activity of naive, non-tolerant cells. This latter observation points to the function of an active, regulatory form of tolerance. As such, we would emphasize that tolerance should not be defined as unresponsiveness since the tolerant state is the consequence of very active immune reactions.
Collapse
Affiliation(s)
- R G Gill
- Barbara Davis Center for Childhood Diabetes, University of Colorado Health Sciences Center, Denver 80262, USA
| | | | | |
Collapse
|
221
|
Abstract
Allograft rejection has been attributed predominantly to alloreactive T cells which recognize intact allogeneic MHC on donor antigen presenting cells (APC). Following transplantation, T cells may also recognize donor derived MHC antigens which have been processed and presented as antigenic peptides in the context of self-MHC by recipient APC--so called indirect allorecognition. This article discusses the possible role of indirect T cell recognition in the graft rejection response with particular emphasis on recent experimental data obtained from in vivo transplant models. Although the relative contribution of indirect allorecognition to the rejection process remains unclear the available evidence suggests that the contribution of self-MHC restricted T cells to graft rejection may have been underestimated.
Collapse
Affiliation(s)
- J A Bradley
- Division of Immunological Science, University of Glasgow, Scotland
| |
Collapse
|
222
|
Terness P, Dufter C, Otto G, Opelz G. Allograft survival following immunization with membrane-bound or soluble peptide MHC class I donor antigens: factors relevant for the induction of rejection by indirect recognition. Transpl Int 1996; 9:2-8. [PMID: 8748404 DOI: 10.1007/bf00336805] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
T cells recognize foreign antigens in the form of peptide fragments resulting from antigen processing by antigen-presenting cells. In contrast to this indirect recognition, MHC molecules of foreign cells can be directly recognized by T cells. Direct recognition has for a long time been considered the only mechanism responsible for transplant rejection. Recent studies have provided evidence of a role of indirect recognition in rejection. In the current series of experiments, we studied the influence of indirect alloactivation, induced either by donor MHC class I peptides or by membrane-bound MHC I molecules, on heart allograft rejection in rats. Recipients were immunized before transplantation with synthetic donor MHC I peptides. The animals developed antibody and T-cell responses. Depending on the rat strain, peptide pretreatment either had no effect on graft survival (DA-->PVG; untreated controls 8.5 +/- 0.6 days, treated rats 9.5 +/- 0.6 days) or led to accelerated rejection (DA-->LEW; untreated controls 7.5 +/- 0.3 days, treated rats 5.1 +/- 0.2 days; P < 0.0002). Importantly, sensitization by indirect activation induced acute rejection in a donor-recipient combination (LEW.1A-->LEW.1WR2) in which neither direct nor indirect recognition led to rejection (untreated controls > 400 days, pretreated rats 15 +/- 4.2 days). Another group of animals was immunized with allogeneic or congenic erythrocytes carrying the MHC I antigen from which the peptides were derived. Although the immunization elicited a measurable immune response, it did not lead to accelerated rejection. We conclude that sensitization by indirect recognition is able to initiate an acute rejection even in recipients in which neither direct nor indirect recognition is effective, and that this effect is strain-dependent. The form in which the donor antigen is administered is decisive for the induction of rejection by indirect activation.
Collapse
Affiliation(s)
- P Terness
- Department of Transplantation Immunology, University of Heidelberg, Germany
| | | | | | | |
Collapse
|
223
|
Abstract
It is now clear from animal studies that indirect allorecognition occurs during allograft rejection and that this pathway plays a role in mediating the rejection process. Whether this pathway is the dominant pathway responsible for chronic rejection remains to be established, but this is an intriguing hypothesis that may have major implications for development of novel therapies for this disorder. In addition, there are data to indicate that indirect allorecognition may play a role in maintenance of allograft acceptance, and that provision of appropriate MHC peptides via different routes can result in potent degrees of specific allo-tolerance. Expanded experimentation in animals and in humans is therefore in order; first, to dissect more closely the molecular basis of allorecognition and establish the role of the indirect pathway in acute versus chronic rejection, and second to utilize the immunomodulatory properties of MHC peptides or other novel strategies which targets indirect allorecognition in promoting graft acceptance.
Collapse
Affiliation(s)
- M H Sayegh
- Laboratory of Immunogenetics and Transplantation, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | |
Collapse
|
224
|
Colovai AI, Molajoni ER, Cortesini R, Suciu-Foca N. New approaches to specific immunomodulation in transplantation. Int Rev Immunol 1996; 13:161-72. [PMID: 8782739 DOI: 10.3109/08830189609061745] [Citation(s) in RCA: 27] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
T cells can recognize foreign MHC antigens by two distinct routes, either directly as intact molecules, or indirectly as processed peptides. Recent evidence strongly suggests that the indirect pathway of allorecognition plays a key role in initiating and sustaining graft rejection. Theoretically, all mismatched HLA alloantigens could generate immunogenic peptides which may be recognized in the context of any of the two self HLA-DR molecules. However, indirect recognition appears to be limited to a single peptide determinant of an allogeneic HLA-DR molecule and restricted by one self HLA-DR molecule. Furthermore, T cells involved in the self-restricted allopeptide recognition express a limited array of T cell receptor variable genes. These findings suggest that selective immune interventions, such as peptide blockade of the self HLA-DR molecule involved in the presentation of the dominant allopeptide, induction of high-zone tolerance or TCR antagonism, may be devised to prevent graft rejection.
Collapse
Affiliation(s)
- A I Colovai
- College of Physicians & Surgeons of Columbia University, Department of Pathology, New York, New York 10032, USA
| | | | | | | |
Collapse
|
225
|
Massard G, Tongio MM, Wihlm JM, Morand G. The dendritic cell lineage: a ubiquitous antigen-presenting organization. Ann Thorac Surg 1996; 61:252-8. [PMID: 8561576 DOI: 10.1016/0003-4975(95)00739-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Dendritic cells are specialized antigen-presenting cells with two unique characteristics: the greatest stimulatory potential and the ability to stimulate naive T-lymphocytes. They originate from the bone marrow and reach their destination via hematogenous or lymphatic migration. Their phenotype is characterized by a high expression of major histocompatibility complex class II molecules and a high expression of adhesion molecules (CD25, CD54, CD58, CD72, and CD80). Pulmonary dendritic cells may be investigated by histologic examination, phenotype analysis, and function studies in a mixed lymphocyte reaction. Their isolation requires enzymatic digestion of lung tissue and subsequent steps of cell separation. The complexity of these manipulations makes it difficult to obtain large numbers of viable cells. A close anatomic relationship with alveolar macrophages underlines a functional interconnection: macrophages down-regulate the antigen-presenting function through release of tumor necrosis factor alpha. Dendritic cells most probably play a major role in lung diseases such as histiocytosis, primary and secondary cancers, and both acute and chronic lung graft rejection. Identification of the precise functional pathways might lead to therapeutic use of modulation of dendritic cell function.
Collapse
Affiliation(s)
- G Massard
- Department of Thoracic Surgery, University Hospital of Strasbourg, France
| | | | | | | |
Collapse
|
226
|
Abstract
A large number of alloantigenic determinants could be generated by both the direct and indirect alloantigen presentation pathways. Hence, a heterogeneous population of T cells expressing a wide variety of receptors would be expected to respond to this diverse array of alloantigenic determinants. However, T cells expressing highly restricted T cell receptor (TCR) variable genes have been reported in a variety of alloimmune responses. A similar phenomenon has been observed in a wide variety of other immune responses, from those induced by superantigens, to very specific responses induced by a single peptide presented by a single MHC molecule. Given this scenario, the limited number of T cell clones which dominate an allograft rejection response, or for that matter an autoimmune response or a tumor specific response, could be therapeutically targeted by virtue of the selected TCR expression.
Collapse
Affiliation(s)
- O J Finn
- Department of Molecular Genetics and Biochemistry, University of Pittsburgh School of Medicine, Pennsylvania 15206, USA
| | | | | |
Collapse
|
227
|
Terness P, Dufter C, Otto G, Opelz G. Allograft survival following immunization with membrane-bound or soluble peptide MHC class I donor antigens: factors relevant for the induction of rejection by indirect recognition. Transpl Int 1996. [DOI: 10.1111/j.1432-2277.1996.tb00845.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
228
|
Zavazava N, Fandrich F, Ott KA, Freese A, Turnewitsch K. Rat MHC class I peptides are immunogenic. Transpl Int 1996; 9 Suppl 1:S337-9. [PMID: 8959859 DOI: 10.1007/978-3-662-00818-8_83] [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: 02/03/2023]
Abstract
We postulated that indirect recognition of MHC-derived peptides might modulate the alloresponse to donor antigen. In this study, we looked at the potential of two class I peptides derived from the alpha 1 and alpha 2 regions of the DA RT1Aa molecule. Lew responder rats were immunized by varying concentrations of two 25mer peptides covering residues 56-81 and 96-120. The injections were under the footpad and were repeated on day 14. The thickness of the footpads was measured to control delayed-type hypersensitivity (DTH). The animals were sacrificed on day 16 and the splenocytes were tested in mixed lymphocyte culture as responders against DA stimulator cells and CAP third-party splenocytes. In addition, the phenotype of the cells was measured using flow cytometry with antibodies against CD4, CD8, CD5, MHC class II, CD25, CD14 and CD19. Peptide 96-120 induced strong sensitization of the Lew recipient animals at concentrations of 200-500 micrograms (n = 4). The stimulation index was 2-3 times higher than that of untreated animals. Peptide 56-81 failed to induce sensitization at the concentrations used, but surprisingly induced a concentration-dependent immunosuppression that was highest at 400 micrograms (n = 4). In proliferation experiments responder Lew rats proliferated only to peptide 56-81 in vitro.
Collapse
Affiliation(s)
- N Zavazava
- Institute of Immunologie, University of Kiel, Germany
| | | | | | | | | |
Collapse
|
229
|
Benichou G, Fedoseyeva EV. The contribution of peptides to T cell allorecognition and allograft rejection. Int Rev Immunol 1996; 13:231-43. [PMID: 8782744 DOI: 10.3109/08830189609061750] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Affiliation(s)
- G Benichou
- Department of Surgery, University of California San Francisco 94143-0508, USA
| | | |
Collapse
|
230
|
Brennan DC, Mohanakumar T, Flye MW. Donor-specific transfusion and donor bone marrow infusion in renal transplantation tolerance: a review of efficacy and mechanisms. Am J Kidney Dis 1995; 26:701-15. [PMID: 7485121 DOI: 10.1016/0272-6386(95)90432-8] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The ultimate goal in transplantation is modulation of the immune response to produce tolerance without immunosuppression. To date only a state of pseudotolerance for the allograft has been achieved through the use of potent pharmacologic and biologic manipulations. Despite these manipulations to prevent acute rejection, chronic rejection eventually results in graft failure. Thus, different strategies have been sought to induce tolerance and prevent acute and chronic rejection. Historically, donor-specific transfusion (DST) was one such strategy attempted. Donor-specific transfusion has been used primarily in living donor organ transplantation. With the concern that DST may sensitize patients, thereby preventing transplantation and introduction of cyclosporine, the use of DST was curtailed. More recently, donor bone marrow (DBM) infusion at the time of cadaveric transplantation has been used to facilitate development of microchimerism and tolerance to abrogate acute and chronic rejection. However, DBM infusion may predispose the recipient to graft-versus-host disease and is not easily accomplished in living donor organ transplantation. The potentially immunomodulating and tolerizing mechanisms of DST and DBM infusion are similar and include induction of anergy, stimulation of anti-anti-HLA antibodies, provision of soluble HLA antigen, suppressor cell and/or veto cell activity, clonal deletion, regulation of cell surface molecules, regulation of cytokines, promotion of microchimerism, or a combination of these. Of these mechanisms, microchimerism with the concomitant persistence of soluble donor HLA antigen is felt by many to be the most important. Although microchimerism is detectable in many patients who are tolerant of their grafts, there is no clear evidence that chimerism is responsible for the induction or maintenance of tolerance.
Collapse
Affiliation(s)
- D C Brennan
- Department of Medicine, Barnes Hospital, Washington University School of Medicine, St Louis, MO 63110, USA
| | | | | |
Collapse
|
231
|
Thomson AW, Lu L, Murase N, Demetris AJ, Rao AS, Starzl TE. Microchimerism, dendritic cell progenitors and transplantation tolerance. Stem Cells 1995; 13:622-39. [PMID: 8590864 PMCID: PMC2963943 DOI: 10.1002/stem.5530130607] [Citation(s) in RCA: 140] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The recent discovery of multilineage donor leukocyte microchimerism in allograft recipients up to three decades after organ transplantation implies the migration and survival of donor stem cells within the host. It has been postulated that in chimeric graft recipients, reciprocal modulation of immune responsiveness between donor and recipient leukocytes may lead, eventually, to the induction of mutual immunologic nonreactivity (tolerance). A prominent donor leukocyte, both in human organ transplant recipients and in animals, has invariably been the bone marrow-derived dendritic cell (DC). These cells have been classically perceived as the most potent antigen-presenting cells but evidence also exists for their tolerogenicity. The liver, despite its comparatively heavy leukocyte content, is the whole organ that is most capable of inducing tolerance. We have observed that DC progenitors propagated from normal mouse liver in response to GM-CSF express only low levels of major histocompatibility complex (MHC) class II antigen and little or no cell surface B7 family T cell costimulatory molecules. They fail to activate resting naive allogeneic T cells. When injected into normal allogeneic recipients, these DC progenitors migrate to T-dependent areas of host lymphoid tissue, where some at least upregulate cell surface MHC class II. These donor-derived cells persist indefinitely, recapitulating the behavior pattern of donor leukocytes after the successful transplantation of all whole organs, but most dramatically after the orthotopic (replacement) engraftment of the liver. A key finding is that in mice, progeny of these donor-derived DC progenitors can be propagated ex vivo from the bone marrow and other lymphoid tissues of nonimmunosuppressed spontaneously tolerant liver allograft recipients. In humans, donor DC can also be grown from the blood of organ allograft recipients whose organ-source chimerism is augmented with donor bone marrow infusion. DC progenitors cannot, however, be propagated from the lymphoid tissue of nonimmunosuppressed cardiac-allografted mice that reject their grafts. These findings are congruent with the possibility that bidirectional leukocyte migration and donor cell chimerism play key roles in acquired transplantation tolerance. Although the cell interactions are undoubtedly complex, a discrete role can be identified for DC under well-defined experimental conditions. Bone marrow-derived DC progenitors (MHC class II+, B7-1dim, B7-2-) induce alloantigen-specific hyporesponsiveness (anergy) in naive T cells in vitro. Moreover, costimulatory molecule-deficient DC progenitors administered systemically prolong the survival of mouse heart or pancreatic islet allografts. How the regulation of donor DC phenotype and function relates to the balance between the immunogenicity and tolerogenicity of organ allografts remains to be determined.
Collapse
Affiliation(s)
- A W Thomson
- Pittsburgh Transplantation Institute, University of Pittsburgh Medical Center, Pennsylvania 15213, USA
| | | | | | | | | | | |
Collapse
|
232
|
Detection of antibodies to HLA1 antigens by enzyme immunoassay. Bull Exp Biol Med 1995. [DOI: 10.1007/bf02445028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
233
|
Bishop DK, DeBruyne LA, Chan S, Xu S, Eichwald EJ. Dissociation of mouse cardiac transplant rejection and donor alloantigen-specific T cell responsiveness. Transpl Immunol 1995; 3:222-8. [PMID: 8581410 DOI: 10.1016/0966-3274(95)80028-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Mouse hearts transplanted into MHC disparate donors are usually rejected 1 week after placement. It is widely accepted that alloantigen-reactive helper T lymphocytes (HTL) and cytotoxic T lymphocytes (CTL) are the key mediators of acute allograft rejection. This report demonstrates that the presence or absence of 'traditional' graft-reactive HTL and CTL is not necessarily related to allograft survival. In these studies, donor/recipient combinations disparate only at MHC or only at minor histocompatibility (mH) loci were employed. Allograft survival was monitored, donor-reactive IL-2 (interleukin-2) producing HTL and CTL were quantified by modified limiting dilution analysis, and serum levels of cytolytic alloantibody were determined. C57BL/10 hearts (H-2b) transplanted into B10.BR (H-2k) recipients (full MHC disparity) enjoyed prolonged survival despite massive infiltration of the allograft by donor-reactive HTL and CTL. IgM, but not IgG, donor-reactive alloantibody was present in the sera of these mice. Hence, traditional IL-2 producing HTL and CTL were not capable of mediating acute graft rejection, nor of providing help for alloantibody isotype switching in this MHC disparate combination. In contrast, C3H/HeN (H-2k) hearts transplanted into B10.BR (H-2k) recipients (mH disparity only) were acutely rejected. Donor-reactive HTL and CTL were rare or not detectable in these recipients, and cytolytic alloantibody was not detectable. Similar observations were made when B10.BR hearts were transplanted into C3H/HeN recipients. Interestingly, treatment of recipients with anti-CD4 monoclonal antibody prevented rejection of mH disparate allografts. Therefore, CD4+ T cells were required for rejection of mH disparate allografts, but this process was independent of detectable IL-2 production or CTL function. Hence, the significance of monitoring 'traditional' T cell functions should be questioned in certain donor/recipient combinations.
Collapse
Affiliation(s)
- D K Bishop
- Department of Surgery, University of Michigan, School of Medicine, Ann Arbor 48109, USA
| | | | | | | | | |
Collapse
|
234
|
Suthanthiran M, Strom TB. Immunobiology and immunopharmacology of organ allograft rejection. J Clin Immunol 1995; 15:161-71. [PMID: 7593462 DOI: 10.1007/bf01541085] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Much has been learned regarding immunobiological mechanisms responsible for the rejection of histoincompatible allografts. There has also been considerable progress in our understanding of mechanisms responsible for tolerance. The new knowledge gained regarding graft destructive alloimmunity process and the mechanisms of action of immunosuppressants have resulted in solid organ graft survival rates that are in excess of 80% at one year posttransplantation. The principles of tolerance mechanism are yet to be successfully applied in the clinic. In this review, molecular and cellular mechanisms of action of clinically useful immunosuppressive drugs are reviewed from the perspective of regulation of the anti-allograft repertory.
Collapse
Affiliation(s)
- M Suthanthiran
- Department of Transplantation and Extracorporeal Therapy, Rogosin Institute, New York Hospital-Cornell Medical Center, New York, USA
| | | |
Collapse
|
235
|
Häyry P. Aspects of allograft rejection, I: Molecular pathology of acute and chronic rejection. Transplant Rev (Orlando) 1995. [DOI: 10.1016/s0955-470x(95)80020-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
236
|
Trauger RJ, Giermakowska W, Wormsley S, Turner J, Jensen FC, Carlo DJ. Autoproliferation in HIV-1-infected patients undergoing active HIV-1-specific immunotherapy. Clin Exp Immunol 1995; 100:7-12. [PMID: 7697926 PMCID: PMC1534259 DOI: 10.1111/j.1365-2249.1995.tb03596.x] [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: 01/26/2023] Open
Abstract
We have observed a treatment-associated autoproliferative response in cultured peripheral blood mononuclear cells (PBMC) of asymptomatic HIV-1-infected subjects receiving a gp120-depleted, inactivated HIV-1 antigen in incomplete Freund's adjuvant (IFA; HIV-1 Immunogen). The frequency and magnitude of the autoproliferative response appeared to be dose-related (P < 0.05), and was not observed in subjects receiving IFA alone. Immunophenotyping of the proliferating cells demonstrated the presence of both CD4+ and CD8+ lymphocytes, with the CD4+ blasts almost exclusively expressing the CD45RO+ phenotype. A comparison of this response with the HIV-1-specific antigen stimulation responses in this cohort revealed a significant correlation between increases in HIV-1-specific cell-mediated immunity and autoproliferation (r2 = 0.61, P < 0.001). These findings suggest that immunization with the HIV-1 Immunogen induces an autoproliferative response that may reflect changes in HIV-1-specific cell-mediated immunity in infected individuals.
Collapse
Affiliation(s)
- R J Trauger
- Immune Response Corporation, Carlsbad, CA, USA
| | | | | | | | | | | |
Collapse
|
237
|
Affiliation(s)
- C J Taylor
- Tissue Typing Laboratory, Addenbrooke's NHS Trust, Cambridge, UK
| | | |
Collapse
|
238
|
Gardner CR. The pharmacology of immunosuppressant drugs in skin transplant rejection in mice and other rodents. GENERAL PHARMACOLOGY 1995; 26:245-71. [PMID: 7590073 DOI: 10.1016/0306-3623(94)00113-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
1. Skin transplantation in rodents is a convenient, widely used method, particularly in mice. It is used as much as an indicator of immune responsiveness as for pharmacological studies. 2. Many differences exist in experimental protocols, both for transplantation and drug administration and in this review, the increase in graft survival time with respect to control times is used to indicate drug effects, in an attempt to account for these differences. 3. The mechanisms underlying skin graft rejection in rodents are described, emphasising the crucial role of both helper and effector T cells. 4. The pharmacology of clinically-used immunosuppressants, including CsA, FK506, rapamycin and purine or pyrimidine synthesis inhibitors, in rodent models of skin transplantation is reviewed. 5. The effects of other potential immunosuppressants and compounds modulating immune responses are described, including the effects of UV light and involvement of platelet-derived factors, prostaglandins and thromboxanes.
Collapse
Affiliation(s)
- C R Gardner
- Roussel Laboratories, Swindon, Wiltshire, England
| |
Collapse
|
239
|
Reinsmoen NL. Posttransplant donor antigen-specific hyporeactivity in human transplantation. Transplant Rev (Orlando) 1995. [DOI: 10.1016/s0955-470x(95)80012-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
240
|
Abstract
The immune system has evolved in the human being as an elaborate mechanism to distinguish itself from all else that is not self. This process serves in the defence against invaders. The cells of the immune system learn to tolerate all tissues, cells and proteins of the body. Failure to control the state of tolerance results in autoimmunity. The understanding of the role of T-cell receptors (TCR), the Major Histocompatibility Complex (MHC), adhesion molecules and growth factors in antigen recognition has lead to the exploration of various means to modulate the immune response. Safety measures exist to prevent the immune system from attacking its host. The antigen has to be recognized by the T-cell. This involves the TCR and the MHC. In addition it must receive a second signal to become activated. The second signal involves a protein such as B7 binding with CD28. Certain specialized cells, macrophages, dendritic cells and activated B-cells can deliver this second signal for activation; receipt of only one signal can prevent activation. The elucidation of the role of cell-to-cell interactions, the adhesion molecules involved and the accessory growth factors provides modalities for selectively modifying the immune response. This would be of great relevance in autoimmunity and transplantation.
Collapse
Affiliation(s)
- K Balakrishnan
- HLA-Laboratory, Hoxworth Blood Center, University of Cincinnati, Ohio 45267, USA
| | | |
Collapse
|
241
|
Abstract
Allogeneic blood transfusion is the most frequent allotransplantation procedure performed on a routine basis with no prior HLA-typing. Roughly 50% of the recipients of unprocessed red cells and platelets become alloimmunized. Evidence also exists for some degree of transfusion-induced immunosuppression. Prior transfusion has been shown to enhance kidney transplant survival and evidence of an increase in tumor recurrence and of infectious complications has also been presented. The presence of donor antigen-presenting cells appears to be a prerequisite for alloimmunization and they must be both viable and capable of presenting a costimulatory signal in order to induce IL-2 secretion and proliferation of responding CD4 T cells. APCs presenting antigen but no costimulatory signal can induce non-responsiveness in CD4 T cells, a possible mechanism of transfusion-induced immunosuppression. APCs in refrigerated blood continue to present antigen but progressively lose their ability to provide costimulation. By day 14 costimulatory capacity is absent and transfusion of such blood should not alloimmunize but could induce some degree of immunosuppression. Further refrigerated storage in excess of 2 to 3 weeks leads to induction of apoptosis in contaminating leukocytes. We have found that alloantigens-expressed on such cells do not appear to be recognized by responder T cells and transfusion of blood stored in excess of 3 weeks should neither alloimmunize nor immunosuppress.
Collapse
Affiliation(s)
- M S Mincheff
- Transplantation Department, Holland Laboratories, American Red Cross, Rockville, MD 20855, USA
| | | |
Collapse
|
242
|
Liu Z, Harris PE, Colovai AI, Reed EF, Maffei A, Suciu-Foca N. Suppression of the indirect pathway of T cell reactivity by high doses of allopeptide. Autoimmunity 1995; 21:173-84. [PMID: 8822275 DOI: 10.3109/08916939509008014] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
T helper cells, which recognize allopeptides processed and presented by self APC, contribute to the generation of both cellular and humoral immune responses against allogeneic transplants. We have explored the hypothesis that the indirect T cell recognition pathway is initiated by soluble MHC antigens and that it can be suppressed by high doses of synthetic peptides corresponding to the dominant alloepitope. T cells from a DR11/7 responder were immunized in vitro with recombinant HLA-DR4 (rDR4). Experiments using partially overlapping synthetic peptides showed that the resulting T cell line (TCL) recognized a single dominant epitope mapping within residues 69-88 of the first domain of the DR4 molecule. In vitro immunization with synthetic allopeptides corresponding to other polymorphic regions, were unable to elicit T cell reactivity against rDR4, although at least one of these peptides (corresponding to residues 13-27) was immunogenic, behaving like a cryptic epitope. The rDR4-specific TCL expressed a limited TCR repertoire and provided help to autologous B cells for the production of specific antibodies. The T cell blastogenic response as well as the transcription and secretion of IL-4 (but not IL-2) was efficiently suppressed by high doses of the dominant allopeptide. These findings support the concept that selective immunointervention of indirect allorecognition can be achieved by use of high doses of antigen or TCR vaccination, as proposed for autoimmune diseases.
Collapse
Affiliation(s)
- Z Liu
- Department of Pathology, College of Physicians and Surgeons of Columbia University, New York 10032, USA
| | | | | | | | | | | |
Collapse
|
243
|
Abstract
The immune defects characterizing infection with the human immunodeficiency virus (HIV) and culminating in the acquired immune deficiency syndrome (AIDS) are the result of a multifactorial disease process, components of which are the occurrence of autoimmune phenomena and opportunistic infection. In this discussion, the observation that both the HIV-1 gp 120 envelope and Mycoplasma genitalium adhesin proteins share an area of significant similarity with the CD4-binding site of the class II major histocompatibility complex (MHC) proteins is placed in this perspective and mechanisms by which interaction within this triad could contribute to the T-cell dysfunction, T-cell depletion, Th1-cell-->Th2-cell shift, B-cell proliferation, hyperglobulinemia and antigen-presenting cell dysfunction observed during the development of AIDS are proposed.
Collapse
Affiliation(s)
- L R Bisset
- Department of Internal Medicine, University Hospital, Zürich, Switzerland
| |
Collapse
|
244
|
|
245
|
Sharma VK, Li B, Khanna A, Sehajpal PK, Suthanthiran M. Which way for drug-mediated immunosuppression? Curr Opin Immunol 1994; 6:784-90. [PMID: 7826535 DOI: 10.1016/0952-7915(94)90085-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The basic immunosuppressive protocol involves the use of multiple drugs, each directed at a discrete site in the T-cell activation cascade. The prevailing paradigm regarding the mechanisms of action of immunosuppressants is that they function to prevent allograft rejection by preventing cell activation, proliferation and/or cytokine production. A new hypothesis is that some of the immunosuppressants might function by stimulating the expression of immunosuppressive molecules and/or cells.
Collapse
Affiliation(s)
- V K Sharma
- Rogsin Institute, Department of Transplantation and Extracorporeal Therapy, New York Hospital-Cornell Medical Center, New York 10021
| | | | | | | | | |
Collapse
|
246
|
Schuurman HJ. Molecular mechanisms of transplant rejection. THE CLINICAL INVESTIGATOR 1994; 72:715-8. [PMID: 7849457 DOI: 10.1007/bf00213000] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
247
|
Wood KJ. Mechanisms of rejection. BAILLIERE'S CLINICAL GASTROENTEROLOGY 1994; 8:425-39. [PMID: 8000091 DOI: 10.1016/0950-3528(94)90029-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- K J Wood
- University of Oxford, Nuffield Department of Surgery, John Radcliffe Hospital, UK
| |
Collapse
|
248
|
Affiliation(s)
- M Suthanthiran
- Rogosin Institute, Department of Medicine, New York Hospital-Cornell Medical Center, New York
| | | |
Collapse
|